1
|
Nakahara T, Fujimoto S, Jinzaki M. Molecular imaging of cardiovascular disease: Current status and future perspective. J Cardiol 2025; 85:386-398. [PMID: 39922562 DOI: 10.1016/j.jjcc.2025.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/15/2025] [Accepted: 01/28/2025] [Indexed: 02/10/2025]
Abstract
Advancements in knowledge of cardiovascular disease, pharmacology, and chemistry have led to the development of newer radiopharmaceuticals and targets for new and more suitable molecules. Molecular imaging encompasses multiple imaging techniques for identifying the characteristics of key components involved in disease. Despite its limitations in spatial resolution, the affinity for key molecules compensates for disadvantages in diagnosing diseases and elucidating their pathophysiology. This review introduce established molecular tracers involved in clinical practice and emerging tracers already applied in clinical studies, classifying the key component in A: artery, specifically those vulnerable plaque (A-I) inflammatory cells [18F-FDG]; A-II) lipid/fatty acid; A-III) hypoxia; A-IV) angiogenesis; A-V) protease [18F/68Ga-FAPI]; A-VI) thrombus/hemorrhage; A-VII) apoptosis and A-VIII) microcalcification [18F-NaF]) and B: myocardium, including myocardial ischemia, infarction and myocardiopathy (B-I) myocardial ischemia; B-II) myocardial infarction (myocardial damage and fibrosis); B-III) myocarditis and endocarditis; B-IV) sarcoidosis; B-V) amyloidosis; B-VI) metabolism; B-VII) innervation imaging). In addition to cardiovascular-specific tracers tested in animal models, many radiotracers may have been developed in other areas, such as oncology imaging or neuroimaging. While this review does not cover all available tracers, some of them hold potential for future use assessing cardiovascular disease. Advances in molecular biology, pharmaceuticals, and imaging sciences will facilitate the identification of precise disease mechanisms, enabling precise diagnoses, better assessment of disease status, and enhanced therapeutic evaluation in this multi-modality era.
Collapse
Affiliation(s)
- Takehiro Nakahara
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan.
| | - Shinichiro Fujimoto
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masahiro Jinzaki
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan
| |
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
|
Thackeray JT. Duplicity of Quantity: Potential and Drawbacks of Cardiac Molecular SPECT Imaging. Circ Cardiovasc Imaging 2024; 17:e017614. [PMID: 39534970 DOI: 10.1161/circimaging.124.017614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Affiliation(s)
- James T Thackeray
- Department of Nuclear Medicine, Translational Cardiovascular Molecular Imaging, Hannover Medical School, Germany
| |
Collapse
|
4
|
Lukovic D, Gyöngyösi M, Pavo IJ, Mester-Tonczar J, Einzinger P, Zlabinger K, Kastner N, Spannbauer A, Traxler D, Pavo N, Goliasch G, Pils D, Jakab A, Szankai Z, Michel-Behnke I, Zhang L, Devaux Y, Graf S, Beitzke D, Winkler J. Increased [ 18F]FDG uptake in the infarcted myocardial area displayed by combined PET/CMR correlates with snRNA-seq-detected inflammatory cell invasion. Basic Res Cardiol 2024; 119:807-829. [PMID: 38922408 PMCID: PMC11461641 DOI: 10.1007/s00395-024-01064-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024]
Abstract
Combined [18F]FDG PET-cardiac MRI imaging (PET/CMR) is a useful tool to assess myocardial viability and cardiac function in patients with acute myocardial infarction (AMI). Here, we evaluated the prognostic value of PET/CMR in a porcine closed-chest reperfused AMI (rAMI) model. Late gadolinium enhancement by PET/CMR imaging displayed tracer uptake defect at the infarction site by 3 days after the rAMI in the majority of the animals (group Match, n = 28). Increased [18F]FDG uptake at the infarcted area (metabolism/contractility mismatch) with reduced tracer uptake in the remote viable myocardium (group Mismatch, n = 12) 3 days after rAMI was observed in the animals with larger infarct size and worse left ventricular ejection fraction (LVEF) (34 ± 8.7 vs 42.0 ± 5.2%), with lower LVEF also at the 1-month follow-up (35.8 ± 9.5 vs 43.0 ± 6.3%). Transcriptome analyses by bulk and single-nuclei RNA sequencing of the infarcted myocardium and border zones (n = 3 of each group, and 3 sham-operated controls) revealed a strong inflammatory response with infiltration of monocytes and macrophages in the infarcted and border areas in Mismatch animals. Our data indicate a high prognostic relevance of combined PET/MRI in the subacute phase of rAMI for subsequent impairment of heart function and underline the adverse effects of an excessive activation of the innate immune system in the initial phase after rAMI.
Collapse
Affiliation(s)
- Dominika Lukovic
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Mariann Gyöngyösi
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria.
| | - Imre J Pavo
- Division of Pediatric Cardiology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Julia Mester-Tonczar
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Patrick Einzinger
- Institute of Information Systems Engineering, Research Unit of Information and Software Engineering, Vienna University of Technology, 1040, Vienna, Austria
| | - Katrin Zlabinger
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Nina Kastner
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Andreas Spannbauer
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Denise Traxler
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Noemi Pavo
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Georg Goliasch
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Dietmar Pils
- Division of General Surgery, Department of Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Andras Jakab
- Center for MR-Research, University Children's Hospital Zurich, Zurich, Switzerland
| | | | - Ina Michel-Behnke
- Division of Pediatric Cardiology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Lu Zhang
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Senta Graf
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Dietrich Beitzke
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Johannes Winkler
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
5
|
Kazimierczyk R, Kaminski KA, Nekolla SG. Cardiac PET/MRI: Recent Developments and Future Aspects. Semin Nucl Med 2024; 54:733-746. [PMID: 38853039 DOI: 10.1053/j.semnuclmed.2024.05.007] [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: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/11/2024]
Abstract
Positron emission tomography/magnetic resonance (PET/MRI) hybrid imaging is now available for over a decade and although the quantity of installed systems is rather low, the number of emerging applications for cardiovascular diseases is still growing. PET/MRI provides integrated images of high quality anatomical and functional assessment obtained by MRI with the possibilities of PET for quantification of molecular parameters such as metabolism, inflammation, and perfusion. In recent years, sequential co-registration of myocardial tissue characterization with its molecular data had become an increasingly helpful tool in clinical practice and an integrated device simplifies this task. This review summarizes recent developments and future possibilities in the use of the PET/MRI in the diagnosis and treatment of cardiovascular disorders.
Collapse
Affiliation(s)
| | - Karol A Kaminski
- Department of Cardiology, Medical University of Bialystok, Bialystok, Poland; Department of Population Medicine and Lifestyle Diseases, Medical University of Bialystok, Bialystok, Poland
| | - Stephan G Nekolla
- Department of Nuclear Medicine, Technical University Munich, Ismaninger Str., Munich, Germany; DZKH (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
6
|
Maier A, Teunissen AJP, Nauta SA, Lutgens E, Fayad ZA, van Leent MMT. Uncovering atherosclerotic cardiovascular disease by PET imaging. Nat Rev Cardiol 2024; 21:632-651. [PMID: 38575752 PMCID: PMC11324396 DOI: 10.1038/s41569-024-01009-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/04/2024] [Indexed: 04/06/2024]
Abstract
Assessing atherosclerosis severity is essential for precise patient stratification. Specifically, there is a need to identify patients with residual inflammation because these patients remain at high risk of cardiovascular events despite optimal management of cardiovascular risk factors. Molecular imaging techniques, such as PET, can have an essential role in this context. PET imaging can indicate tissue-based disease status, detect early molecular changes and provide whole-body information. Advances in molecular biology and bioinformatics continue to help to decipher the complex pathogenesis of atherosclerosis and inform the development of imaging tracers. Concomitant advances in tracer synthesis methods and PET imaging technology provide future possibilities for atherosclerosis imaging. In this Review, we summarize the latest developments in PET imaging techniques and technologies for assessment of atherosclerotic cardiovascular disease and discuss the relationship between imaging readouts and transcriptomics-based plaque phenotyping.
Collapse
Affiliation(s)
- Alexander Maier
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Abraham J P Teunissen
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sheqouia A Nauta
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Esther Lutgens
- Cardiovascular Medicine and Immunology, Experimental Cardiovascular Immunology Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Zahi A Fayad
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mandy M T van Leent
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
7
|
Saric P, Bois JP, Giudicessi JR, Rosenbaum AN, Kusmirek JE, Lin G, Chareonthaitawee P. Imaging of Cardiac Sarcoidosis: An Update and Future Aspects. Semin Nucl Med 2024; 54:701-716. [PMID: 38480041 DOI: 10.1053/j.semnuclmed.2024.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 02/16/2024] [Accepted: 02/19/2024] [Indexed: 08/20/2024]
Abstract
Cardiac sarcoidosis (CS), an increasingly recognized disease of unknown etiology, is associated with significant morbidity and mortality. Given the limited diagnostic yield of traditional endomyocardial biopsy (EMB), there is increasing reliance on multimodality cardiovascular imaging in the diagnosis and management of CS, with EMB being largely supplanted by the use of 18F-fluorodeoxyglucose (FDG-PET) and cardiac magnetic resonance imaging (CMR). This article aims to provide a comprehensive review of imaging modalities currently utilized in the screening, diagnosis, and monitoring of CS, while highlighting the latest developments in each area.
Collapse
Affiliation(s)
- Petar Saric
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - John P Bois
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | | | | | | | - Grace Lin
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | | |
Collapse
|
8
|
Saraste A, Ståhle M, Roivainen A, Knuuti J. Molecular Imaging of Heart Failure: An Update and Future Trends. Semin Nucl Med 2024; 54:674-685. [PMID: 38609753 DOI: 10.1053/j.semnuclmed.2024.03.005] [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: 03/12/2024] [Accepted: 03/19/2024] [Indexed: 04/14/2024]
Abstract
Molecular imaging can detect and quantify pathophysiological processes underlying heart failure, complementing evaluation of cardiac structure and function with other imaging modalities. Targeted tracers have enabled assessment of various cellular and subcellular mechanisms of heart failure aiming for improved phenotyping, risk stratification, and personalized therapy. This review outlines the current status of molecular imaging in heart failure, accompanied with discussion on novel developments. The focus is on radionuclide methods with data from clinical studies. Imaging of myocardial metabolism can identify left ventricle dysfunction caused by myocardial ischemia that may be reversible after revascularization in the presence of viable myocardium. In vivo imaging of active inflammation and amyloid deposition have an established role in the detection of cardiac sarcoidosis and transthyretin amyloidosis. Innervation imaging has well documented prognostic value in predicting heart failure progression and arrhythmias. Tracers specific for inflammation, angiogenesis and myocardial fibrotic activity are in earlier stages of development, but have demonstrated potential value in early characterization of the response to myocardial injury and prediction of cardiac function over time. Early detection of disease activity is a key for transition from medical treatment of clinically overt heart failure towards a personalized approach aimed at supporting repair and preventing progressive cardiac dysfunction.
Collapse
Affiliation(s)
- Antti Saraste
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland; Heart Center, Turku University Hospital and University of Turku, Turku, Finland.
| | - Mia Ståhle
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
| | - Anne Roivainen
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
| | - Juhani Knuuti
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
| |
Collapse
|
9
|
Garg M, Gandhi K, Gera P, Jadhav SM, Mohanty B, Gurjar M, Sandupatla B, Gala R, Chaudhari P, Prasad M, Chinnaswamy G, Gota V. Implications of chronic moderate protein-deficiency malnutrition on doxorubicin pharmacokinetics and cardiotoxicity in early post-weaning stage. Life Sci 2024; 350:122765. [PMID: 38830506 DOI: 10.1016/j.lfs.2024.122765] [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: 12/17/2023] [Revised: 03/14/2024] [Accepted: 05/29/2024] [Indexed: 06/05/2024]
Abstract
BACKGROUND Malnutrition is a common problem in developing countries, and the impact of severe malnutrition on optimal treatment outcomes of chemotherapy in pediatric cancer patients is well documented. However, despite being a more prevalent and distinct entity, moderate malnutrition is until now unexplored for its effects on treatment outcomes. AIMS In this study we aimed to investigate the molecular basis of altered pharmacokinetics and cardiotoxicity of doxorubicin observed in early-life chronic moderate protein deficiency malnutrition. MATERIALS AND METHODS We developed an animal model of early-life moderate protein-deficiency malnutrition and validated it using clinical samples. This model was used to study pharmacokinetic and toxicity changes and was further utilized to study the molecular changes in liver and heart to get mechanistic insights. KEY FINDINGS Here we show that moderate protein-deficiency malnutrition in weanling rats causes changes in drug disposition in the liver by modification of hepatic ABCC3 and MRP2 transporters through the TNFα signalling axis. Furthermore, malnourished rats in repeat-dose doxorubicin toxicity study showed higher toxicity and mortality. A higher accumulation of doxorubicin in the heart was observed which was associated with alterations in cardiac metabolic pathways and increased cardiotoxicity. SIGNIFICANCE Our findings indicate that moderate malnutrition causes increased susceptibility towards toxic side effects of chemotherapy. These results may necessitate further investigations and new guidelines on the dosing of chemotherapy in moderately malnourished pediatric cancer patients.
Collapse
Affiliation(s)
- Megha Garg
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research, and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra 410210, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra 400094, India
| | - Khushboo Gandhi
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research, and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra 410210, India
| | - Poonam Gera
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra 400094, India; Biorepository, Advanced Centre for Treatment Research and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra 410210, India
| | - Shraddha Mahesh Jadhav
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research, and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra 410210, India
| | - Bhabani Mohanty
- Department of Comparative Oncology and Small Animal Imaging Facility, ACTREC, Tata Memorial Center, Kharghar, Navi-Mumbai, Maharashtra 410210, India
| | - Murari Gurjar
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research, and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra 410210, India; Department of Clinical Pharmacology, Mahamana Pandit Madan Mohan Malviya Cancer Centre, Banaras Hindu University Campus, Varanasi, Uttar Pradesh 221005, India
| | | | - Rajul Gala
- Paediatric Oncology, Tata Memorial Hospital, Mumbai, India
| | - Pradip Chaudhari
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra 400094, India; Department of Comparative Oncology and Small Animal Imaging Facility, ACTREC, Tata Memorial Center, Kharghar, Navi-Mumbai, Maharashtra 410210, India
| | - Maya Prasad
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra 400094, India; Paediatric Oncology, Tata Memorial Hospital, Mumbai, India
| | - Girish Chinnaswamy
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra 400094, India; Paediatric Oncology, Tata Memorial Hospital, Mumbai, India
| | - Vikram Gota
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research, and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra 410210, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra 400094, India.
| |
Collapse
|
10
|
Diekmann J, Thackeray JT. Strength in numbers: Advancing the clinical reality fibroblast activation protein imaging in cardiovascular disease. J Nucl Cardiol 2024; 38:102016. [PMID: 39098648 DOI: 10.1016/j.nuclcard.2024.102016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/06/2024]
Affiliation(s)
- Johanna Diekmann
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany.
| | - James T Thackeray
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| |
Collapse
|
11
|
Sazonova SI, Ilushenkova JN. Imaging of postinfarct inflammation. J Nucl Cardiol 2024; 35:101808. [PMID: 38244978 DOI: 10.1016/j.nuclcard.2024.101808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 01/10/2024] [Indexed: 01/22/2024]
Affiliation(s)
- Svetlana I Sazonova
- Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russian Federation.
| | - Julia N Ilushenkova
- Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russian Federation
| |
Collapse
|
12
|
Thackeray JT, Lavine KJ, Liu Y. Imaging Inflammation Past, Present, and Future: Focus on Cardioimmunology. J Nucl Med 2023; 64:39S-48S. [PMID: 37918845 DOI: 10.2967/jnumed.122.264865] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 09/12/2023] [Indexed: 11/04/2023] Open
Abstract
Growing evidence implicates the immune system as a critical mediator of cardiovascular disease progression and a viable therapeutic target. Increased inflammatory cell activity is seen in the full spectrum of disorders from early-stage atherosclerosis through myocardial infarction, cardiomyopathy, and chronic heart failure. Although therapeutic strategies to modulate inflammation have shown promise in preclinical animal models, efficacy in patients has been modest owing in part to the variable severity of inflammation across individuals. The diverse leukocyte subpopulations involved in different aspects of heart disease pose a challenge to effective therapy, wherein adverse and beneficial aspects of inflammation require appropriate balance. Noninvasive molecular imaging enables tissue-level interrogation of inflammatory cells in the heart and vasculature to provide mechanistic and temporal insights into disease progression. Although clinical imaging has relied on 18F-FDG as a nonselective and crude marker of inflammatory cell activity, new imaging probes targeting cell surface markers of different leukocyte subpopulations present the opportunity to visualize and quantify distinct phases of cardiac and vessel wall inflammation. Similarly, therapies are evolving to more effectively isolate adverse from beneficial cell populations. This parallel development of immunocardiology and molecular imaging provides the opportunity to refine treatments using imaging guidance, building toward mechanism-based precision medicine. Here, we discuss progress in molecular imaging of immune cells in cardiology from use of 18F-FDG in the past to the present expansion of the radiotracer arsenal and then to a future theranostic paradigm of tracer-therapy compound pairs with shared targets. We then highlight the critical experiments required to advance the field from preclinical concept to clinical reality.
Collapse
Affiliation(s)
- James T Thackeray
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany;
| | - Kory J Lavine
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; and
| | - Yongjian Liu
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
13
|
Fukushima K, Ito H, Takeishi Y. Comprehensive assessment of molecular function, tissue characterization, and hemodynamic performance by non-invasive hybrid imaging: Potential role of cardiac PETMR. J Cardiol 2023; 82:286-292. [PMID: 37343931 DOI: 10.1016/j.jjcc.2023.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/16/2023] [Accepted: 05/23/2023] [Indexed: 06/23/2023]
Abstract
Noninvasive cardiovascular imaging plays a key role in diagnosis and patient management including monitoring treatment efficacy. The usefulness of noninvasive cardiovascular imaging has been extensively studied and shown to have high diagnostic reliability and prognostic significance, while the nondiagnostic results frequently encountered with single imaging modality require complementary or alternative imaging techniques. Hybrid cardiac imaging was initially introduced to integrate anatomical and functional information to enhance the diagnostic performance, and lately employed as a strategy for comprehensive assessment of the underlying pathophysiology of diseases. More recently, the utility of computed tomography has grown in diversity, and emerged from being an exploratory technique allowing functional measurement such as stress dynamic perfusion. Cardiac magnetic resonance imaging (CMR) is widely accepted as a robust tool for evaluation of cardiac function, fibrosis, and edema, yielding high spatial resolution and soft-tissue contrast. However, the use of intravenous contrast materials is typically required for accurate diagnosis with these imaging modalities, despite the associated risk of renal toxicity. Nuclear cardiology, established as a molecular imaging technique, has advantages in visualization of the disease-specific biological process at cellular level using numerous probes without requiring contrast materials. Various imaging modalities should be appropriately used sequentially to assess concomitant disease and the progression over time. Therefore, simultaneous evaluation combining high spatial resolution and disease-specific imaging probe is a useful approach to identify the regional activity and the stage of the disease. Given the recent advance and potential of multiparametric CMR and novel nuclide tracers, hybrid positron emission tomography MR is becoming an ideal tool for disease-specific imaging.
Collapse
Affiliation(s)
- Kenji Fukushima
- Department of Radiology and Nuclear Medicine, Fukushima Medical University, Fukushima, Japan.
| | - Hiroshi Ito
- Department of Radiology and Nuclear Medicine, Fukushima Medical University, Fukushima, Japan
| | - Yasuchika Takeishi
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
14
|
Lavine KJ, Sultan D, Luehmann H, Detering L, Zhang X, Heo GS, Zhang X, Hoelscher M, Harrison K, Combadière C, Laforest R, Kreisel D, Woodard PK, Brody SL, Gropler RJ, Liu Y. CCR2 Imaging in Human ST-Segment Elevation Myocardial Infarction. NATURE CARDIOVASCULAR RESEARCH 2023; 2:874-880. [PMID: 38947883 PMCID: PMC11210834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Among the diverse populations of myeloid cells that reside within the healthy and diseased heart, C-C chemokine receptor 2 (CCR2) is specifically expressed on inflammatory populations of monocytes and macrophages that contribute to the development and progression of heart failure1-4. Here, we evaluated a peptide-based imaging probe (64Cu-DOTA-ECL1i) that specifically recognizes CCR2+ monocytes and macrophages for human cardiac imaging. Compared to healthy controls, 64Cu-DOTA-ECL1i heart uptake was increased in subjects following acute myocardial infarction, predominately localized within the infarct area, and was associated with impaired myocardial wall motion. These findings establish the feasibility of molecular imaging of CCR2 expression to visualize inflammatory monocytes and macrophages in the injured human heart.
Collapse
Affiliation(s)
- Kory J Lavine
- Department of Medicine, Washington University School of Medicine, St. Louis, MO USA
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO USA
- Department of Immunology and Pathology, Washington University School of Medicine, St. Louis, MO USA
| | - Deborah Sultan
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Hannah Luehmann
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Lisa Detering
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Xiaohui Zhang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Gyu Seong Heo
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Xiuli Zhang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Michelle Hoelscher
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Kitty Harrison
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Christophe Combadière
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, Cimi-Paris, F-75013 Paris, France
| | - Richard Laforest
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Daniel Kreisel
- Department of Immunology and Pathology, Washington University School of Medicine, St. Louis, MO USA
- Department of Surgery, Washington University School of Medicine, St. Louis, MO USA
| | - Pamela K. Woodard
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Steven L. Brody
- Department of Medicine, Washington University School of Medicine, St. Louis, MO USA
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Robert J Gropler
- Department of Medicine, Washington University School of Medicine, St. Louis, MO USA
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Yongjian Liu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO USA
| |
Collapse
|
15
|
Saraste A, Knuuti J, Bengel F. Phenotyping heart failure by nuclear imaging of myocardial perfusion, metabolism, and molecular targets. Eur Heart J Cardiovasc Imaging 2023; 24:1318-1328. [PMID: 37294318 PMCID: PMC10531130 DOI: 10.1093/ehjci/jead128] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 05/26/2023] [Indexed: 06/10/2023] Open
Abstract
Nuclear imaging techniques can detect and quantify pathophysiological processes underlying heart failure, complementing evaluation of cardiac structure and function with other imaging modalities. Combined imaging of myocardial perfusion and metabolism can identify left ventricle dysfunction caused by myocardial ischaemia that may be reversible after revascularization in the presence of viable myocardium. High sensitivity of nuclear imaging to detect targeted tracers has enabled assessment of various cellular and subcellular mechanisms of heart failure. Nuclear imaging of active inflammation and amyloid deposition is incorporated into clinical management algorithms of cardiac sarcoidosis and amyloidosis. Innervation imaging has well-documented prognostic value with respect to heart failure progression and arrhythmias. Emerging tracers specific for inflammation and myocardial fibrotic activity are in earlier stages of development but have demonstrated potential value in early characterization of the response to myocardial injury and prediction of adverse left ventricular remodelling. Early detection of disease activity is a key for transition from broad medical treatment of clinically overt heart failure towards a personalized approach aimed at supporting repair and preventing progressive failure. This review outlines the current status of nuclear imaging in phenotyping heart failure and combines it with discussion on novel developments.
Collapse
Affiliation(s)
- Antti Saraste
- Turku PET Centre, Turku University Hospital and University of Turku, Kiinamyllynkatu 4–8, 20520 Turku, Finland
- Heart Center, Turku University Hospital and University of Turku, Hämeentie 11, 20520 Turku, Finland
| | - Juhani Knuuti
- Turku PET Centre, Turku University Hospital and University of Turku, Kiinamyllynkatu 4–8, 20520 Turku, Finland
| | - Frank Bengel
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| |
Collapse
|
16
|
Thorp EB. Cardiac macrophages and emerging roles for their metabolism after myocardial infarction. J Clin Invest 2023; 133:e171953. [PMID: 37712418 PMCID: PMC10503791 DOI: 10.1172/jci171953] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023] Open
Abstract
Interest in cardioimmunology has reached new heights as the experimental cardiology field works to tap the unrealized potential of immunotherapy for clinical care. Within this space is the cardiac macrophage, a key modulator of cardiac function in health and disease. After a myocardial infarction, myeloid macrophages both protect and harm the heart. To varying degrees, such outcomes are a function of myeloid ontogeny and heterogeneity, as well as functional cellular plasticity. Diversity is further shaped by the extracellular milieu, which fluctuates considerably after coronary occlusion. Ischemic limitation of nutrients constrains the metabolic potential of immune cells, and accumulating evidence supports a paradigm whereby macrophage metabolism is coupled to divergent inflammatory consequences, although experimental evidence for this in the heart is just emerging. Herein we examine the heterogeneous cardiac macrophage response following ischemic injury, with a focus on integrating putative contributions of immunometabolism and implications for therapeutically relevant cardiac injury versus cardiac repair.
Collapse
|
17
|
DeBerge M, Chaudhary R, Schroth S, Thorp EB. Immunometabolism at the Heart of Cardiovascular Disease. JACC Basic Transl Sci 2023; 8:884-904. [PMID: 37547069 PMCID: PMC10401297 DOI: 10.1016/j.jacbts.2022.12.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/21/2022] [Accepted: 12/27/2022] [Indexed: 08/08/2023]
Abstract
Immune cell function among the myocardium, now more than ever, is appreciated to regulate cardiac function and pathophysiology. This is the case for both innate immunity, which includes neutrophils, monocytes, dendritic cells, and macrophages, as well as adaptive immunity, which includes T cells and B cells. This function is fueled by cell-intrinsic shifts in metabolism, such as glycolysis and oxidative phosphorylation, as well as metabolite availability, which originates from the surrounding extracellular milieu and varies during ischemia and metabolic syndrome. Immune cell crosstalk with cardiac parenchymal cells, such as cardiomyocytes and fibroblasts, is also regulated by complex cellular metabolic circuits. Although our understanding of immunometabolism has advanced rapidly over the past decade, in part through valuable insights made in cultured cells, there remains much to learn about contributions of in vivo immunometabolism and directly within the myocardium. Insight into such fundamental cell and molecular mechanisms holds potential to inform interventions that shift the balance of immunometabolism from maladaptive to cardioprotective and potentially even regenerative. Herein, we review our current working understanding of immunometabolism, specifically in the settings of sterile ischemic cardiac injury or cardiometabolic disease, both of which contribute to the onset of heart failure. We also discuss current gaps in knowledge in this context and therapeutic implications.
Collapse
Affiliation(s)
| | | | - Samantha Schroth
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Edward B. Thorp
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
18
|
Whittington B, Dweck MR, van Beek EJR, Newby D, Williams MC. PET-MRI of Coronary Artery Disease. J Magn Reson Imaging 2023; 57:1301-1311. [PMID: 36524452 DOI: 10.1002/jmri.28554] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 12/23/2022] Open
Abstract
Simultaneous positron emission tomography and magnetic resonance imaging (PET-MRI) combines the anatomical detail and tissue characterization of MRI with the functional information from PET. Within the coronary arteries, this hybrid technique can be used to identify biological activity combined with anatomically high-risk plaque features to better understand the processes underlying coronary atherosclerosis. Furthermore, the downstream effects of coronary artery disease on the myocardium can be characterized by providing information on myocardial perfusion, viability, and function. This review will describe the current capabilities of PET-MRI in coronary artery disease and discuss the limitations and future directions of this emerging technique. LEVEL OF EVIDENCE: 5 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Beth Whittington
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging Facility QMRI, University of Edinburgh, Edinburgh, UK
| | - Marc R Dweck
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging Facility QMRI, University of Edinburgh, Edinburgh, UK
| | | | - David Newby
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging Facility QMRI, University of Edinburgh, Edinburgh, UK
| | - Michelle C Williams
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging Facility QMRI, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
19
|
Lang CI, Dahmen A, Vasudevan P, Lemcke H, Gäbel R, Öner A, Ince H, David R, Wolfien M. Cardiac cell therapies for the treatment of acute myocardial infarction in mice: systematic review and meta-analysis. Cytotherapy 2023; 25:640-652. [PMID: 36890093 DOI: 10.1016/j.jcyt.2023.01.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/22/2023] [Accepted: 01/24/2023] [Indexed: 03/08/2023]
Abstract
Backgound Aims: This meta-analysis aims at summarizing the whole body of research on cell therapies for acute myocardial infarction (MI) in the mouse model to bring forward ongoing research in this field of regenerative medicine. Despite rather modest effects in clinical trials, pre-clinical studies continue to report beneficial effects of cardiac cell therapies for cardiac repair following acute ischemic injury. Results: The authors' meta-analysis of data from 166 mouse studies comprising 257 experimental groups demonstrated a significant improvement in left ventricular ejection fraction of 10.21% after cell therapy compared with control animals. Subgroup analysis indicated that second-generation cell therapies such as cardiac progenitor cells and pluripotent stem cell derivatives had the highest therapeutic potential for minimizing myocardial damage post-MI. Conclusions: Whereas the vision of functional tissue replacement has been replaced by the concept of regional scar modulation in most of the investigated studies, rather basic methods for assessing cardiac function were most frequently used. Hence, future studies will highly benefit from integrating methods for assessment of regional wall properties to evolve a deeper understanding of how to modulate cardiac healing after acute MI.
Collapse
Affiliation(s)
| | - Anika Dahmen
- Department of Cardiac Surgery, Rostock University Medical Center, Rostock, Germany; Department of Life, Light and Matter, University of Rostock, Rostock, Germany
| | - Praveen Vasudevan
- Department of Cardiac Surgery, Rostock University Medical Center, Rostock, Germany; Department of Life, Light and Matter, University of Rostock, Rostock, Germany
| | - Heiko Lemcke
- Department of Cardiac Surgery, Rostock University Medical Center, Rostock, Germany; Department of Life, Light and Matter, University of Rostock, Rostock, Germany
| | - Ralf Gäbel
- Department of Cardiac Surgery, Rostock University Medical Center, Rostock, Germany; Department of Life, Light and Matter, University of Rostock, Rostock, Germany
| | - Alper Öner
- Department of Cardiology, Rostock University Medical Center, Rostock, Germany
| | - Hüseyin Ince
- Department of Cardiology, Rostock University Medical Center, Rostock, Germany
| | - Robert David
- Department of Cardiac Surgery, Rostock University Medical Center, Rostock, Germany; Department of Life, Light and Matter, University of Rostock, Rostock, Germany
| | - Markus Wolfien
- Institute of Medical Informatics and Biometry, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
20
|
Heo GS, Diekmann J, Thackeray JT, Liu Y. Nuclear Methods for Immune Cell Imaging: Bridging Molecular Imaging and Individualized Medicine. Circ Cardiovasc Imaging 2023; 16:e014067. [PMID: 36649445 PMCID: PMC9858352 DOI: 10.1161/circimaging.122.014067] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Inflammation is a key mechanistic contributor to the progression of cardiovascular disease, from atherosclerosis through ischemic injury and overt heart failure. Recent evidence has identified specific roles of immune cell subpopulations in cardiac pathogenesis that diverges between individual patients. Nuclear imaging approaches facilitate noninvasive and serial quantification of inflammation severity, offering the opportunity to predict eventual outcome, stratify patient risk, and guide novel targeted molecular therapies against specific leukocyte subpopulations. Here, we will discuss the established and emerging nuclear imaging methods to label and track exogenous and endogenous immune cells, with a particular focus on clinical situations in which targeted molecular inflammation imaging would be advantageous. The expanding options for imaging inflammation provide the foundation to bridge between molecular imaging and individual therapy.
Collapse
Affiliation(s)
- Gyu Seong Heo
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO (G.S.H., Y. L.)
| | - Johanna Diekmann
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany (J.D., J.T.T.)
| | - James T Thackeray
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany (J.D., J.T.T.)
| | - Yongjian Liu
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO (G.S.H., Y. L.)
| |
Collapse
|
21
|
Wienecke LM, Leid JM, Leuschner F, Lavine KJ. Imaging Targets to Visualize the Cardiac Immune Landscape in Heart Failure. Circ Cardiovasc Imaging 2023; 16:e014071. [PMID: 36649453 PMCID: PMC9858350 DOI: 10.1161/circimaging.122.014071] [Citation(s) in RCA: 6] [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: 01/19/2023]
Abstract
Heart failure involves a complex interplay between diverse populations of immune cells that dynamically shift across the natural history of disease. Within this context, the character of the immune response is a key determinant of clinical outcomes. Recent technological advances in single-cell transcriptomic, spatial, and proteomic technologies have fueled an explosion of new and clinically relevant insights into distinct immune cell populations that reside within the diseased heart including potential targets for molecular imaging and therapy. In this review, we will discuss the immune cell types and their respective functions with respect to myocardial infarction remodeling, dilated cardiomyopathy, and heart failure with preserved ejection fraction. In addition, we give a brief overview regarding myocarditis and cardiac sarcoidosis as inflammatory heart failure etiologies. We will highlight markers and cell populations as targets for molecular imaging to visualize inflammation and tissue healing and discuss clinical implications including the development and implementation of precision medicine approaches.
Collapse
Affiliation(s)
- Laura M. Wienecke
- Department of Cardiology, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg, Heidelberg, Germany
| | - Jamison M. Leid
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Florian Leuschner
- Department of Cardiology, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg, Heidelberg, Germany
| | - Kory J. Lavine
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Center for Regenerative Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| |
Collapse
|
22
|
Xi XY, Liu Z, Wang LF, Yang MF. Prognostic value of cardiac inflammation in ST-segment elevation myocardial infarction: A 18F-fluorodeoxyglucose PET/CT study. J Nucl Cardiol 2022; 29:3018-3027. [PMID: 34773185 DOI: 10.1007/s12350-021-02858-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 10/24/2021] [Indexed: 01/18/2023]
Abstract
BACKGROUND 18F-fluorodeoxyglucose (FDG) imaging is used to detect cardiac inflammation and predict functional outcome in acute myocardial infarction (MI). However, data on the correlation of post-MI acute cardiac inflammation evaluated by 18F-FDG imaging and major adverse cardiac events (MACE) are limited. Therefore, we sought to explore the prognostic value of cardiac 18F-FDG imaging in patients with acute ST-segment elevation MI (STEMI). METHODS Thirty-six patients with STEMI underwent 18F-FDG positron emission tomography/computed tomography (PET/CT) 5 days after primary percutaneous coronary intervention. 18F-FDG activity in infarcted and remote regions, as well as peri-coronary adipose tissue (PCAT), were measured and expressed as the maximum standardized uptake value (SUVmax). Patients were followed to determine the occurrence of MACE. RESULTS The infarcted myocardium had a higher 18F-FDG intensity than the remote area. Moreover, the PCAT of culprit coronary arteries showed a higher 18F-FDG uptake than that of non-culprit arteries. Multivariate Cox regression analysis showed that increased SUVmax of PCAT [HR 5.198; 95% CI (1.058, 25.537), P = .042] was independently associated with a higher risk of MACE. CONCLUSIONS Enhanced PCAT activity after acute MI is related to the occurrence of MACE, and 18F-FDG PET/CT plays a promising role in providing prognostic information in patients with STEMI.
Collapse
Affiliation(s)
- Xiao-Ying Xi
- Department of Nuclear Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Ze Liu
- Department of Cardiology, Peking University Third Hospital Yanqing Hospital, Beijing, 102100, China
| | - Le-Feng Wang
- Center of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Min-Fu Yang
- Department of Nuclear Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
23
|
Hua C, Cai Q, Xi XY, Lin M, Wang L, Li L, Yao D, Liu X, Zhao L, Wang L, Su P, Xie B. 99mTc-sestamibi and 18F-fluorodeoxyglucose imaging in patients with cardiogenic shock: A pilot study. Front Cardiovasc Med 2022; 9:1047577. [DOI: 10.3389/fcvm.2022.1047577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 10/19/2022] [Indexed: 11/10/2022] Open
Abstract
BackgroundWhether perfusion/metabolism imaging differs between matched ST-segment elevation myocardial infarction (STEMI) patients with and without cardiogenic shock (CS) remains unknown.MethodsSeventeen STEMI patients with CS (13 men, 60 ± 12 years) and 16 matched STEMI patients without CS (15 men, 54 ± 15 years) were prospectively recruited. All patients underwent baseline 99mTc-sestamibi/18F-fluorodeoxyglucose (FDG) imaging and echocardiography 6 ± 2 days post-infarction. Nine patients with CS and seven without CS had repeated imaging 98 ± 7 days post-infarction. The total perfusion deficit (TPD) and total FDG uptake deficit (TFD) were calculated to assess the percentages of impaired perfusion and metabolism over the left ventricle. Patients were followed up for 337 days (213–505 days) and the major adverse cardiac events (MACE) were recorded.ResultsTPD was greater in patient with CS and was independently related to the presence of CS (OR: 4.36, p = 0.013). Both acute- and convalescent TFD were inversely related to the improvement ratio of LVEF (r-values: −0.62, −0.73; both p < 0.05). MACE occurred in 16 patients (10 CS and 6 non-CS), and acute TFD was predictive of MACE in those with CS (HR: 2.06, p = 0.038).ConclusionIn this pilot study, we demonstrated that STEMI patients with CS had a significantly increased TPD, which was relevant to the presence of CS. Acute TFD was associated with improvement in LVEF, and was predictive of MACE in patients with CS.
Collapse
|
24
|
Rischpler C, Seifert R. Combined PET and MRI for the masses! : At least for the cardiac ones. J Nucl Cardiol 2022; 29:1518-1519. [PMID: 34935109 PMCID: PMC9351608 DOI: 10.1007/s12350-021-02881-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 10/31/2021] [Indexed: 11/15/2022]
Affiliation(s)
- Christoph Rischpler
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| | - Robert Seifert
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
25
|
Su H, Lu D, Shen M, Feng L, Xu C. Evaluating the cardioprotective effect of metformin on myocardial ischemia-reperfusion injury using dynamic 18F-FDG micro-PET/CT imaging. BMC Cardiovasc Disord 2022; 22:310. [PMID: 35811313 PMCID: PMC9272551 DOI: 10.1186/s12872-022-02750-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/06/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND The molecular mechanisms of protective effect of metformin (Met) on ischemic myocardium have not been fully understood. This study aims to evaluate the cardioprotective effect of metformin on myocardial ischemia-reperfusion injury (MIRI) in rat models at different time points using dynamic 18F-FDG micro-PET/CT imaging. METHODS The I/R injury model in SD rats was established by ligation of left anterior descending coronary artery near the pulmonary arch root for 30 min. SD rats (n = 12) were randomly divided into 2 groups: Control group (n = 6) without any intervention and Met group (n = 6) with oral administration of metformin (50 mg/kg) twice a day. Gated 18F-FDG (40Mbq) micro-PET/CT imaging was performed for 10 min at different time points (day 1st, day 7th, day 14th and day 30th after operation). Volumes of interest were drawn to identify different myocardium regions (ischemia center, peri-ischemia area and remote area). Standardized uptake values (SUVs) (SUVmean and SUVmax) were analyzed to evaluate the FDG uptake activity, and then the center/remote ratio was calculated. In addition, the left ventricular (LV) end-diastolic volume (EDV), end-systolic volume (ESV) and LV ejection fraction (LVEF) were obtained. On the 30th day, all rats were scarified and myocardial ischemia was analyzed by HE staining and confirmed by pathology. RESULTS In the Control group, the center/remote ratio showed no obvious change trend at each time point after reperfusion, while the LV EDV increased gradually over time, and they were significantly negatively correlated (r = - 0.507, p < 0.05). In the Met group, the center/remote ratio gradually increased with time, there was no significant correlation between center/remote ratio and LV EDV (r = - 0.078, p > 0.05). On the 30th day, the center/remote ratio of the Met group was significantly higher than that of the Control group (0.81 ± 0.06 vs. 0.65 ± 0.09, p < 0.05), while LV EDV in Met group was significantly lower than in Control group (358.21 ± 22.62 vs. 457.53 ± 29.91, p < 0.05). There was no significant difference of LVEF between Met group and Control group at different time points after reperfusion (p < 0.05). HE staining showed that the myocardial infarction and fibrosis in ischemic center area of the Control group was more serious than that of the Met group. CONCLUSIONS Met could attenuate the severity of MIRI, delay and prevent the progress of LV remodeling. The cardioprotective progress could be dynamically assessed by 18F-FDG micro-PET/CT imaging.
Collapse
Affiliation(s)
- Hang Su
- Department of Nuclear Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Diyu Lu
- Department of Nuclear Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Mingkui Shen
- School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Avenue, Nanshan District, Shenzhen, 518055, People's Republic of China
| | - Li Feng
- School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Avenue, Nanshan District, Shenzhen, 518055, People's Republic of China
| | - Chuangye Xu
- School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Avenue, Nanshan District, Shenzhen, 518055, People's Republic of China.
| |
Collapse
|
26
|
Stendahl JC, Liu Z, Boutagy NE, Nataneli E, Daghighian F, Sinusas AJ. Prototype device for endoventricular beta-emitting radiotracer detection and molecularly-guided intervention. J Nucl Cardiol 2022; 29:663-676. [PMID: 32820423 PMCID: PMC7895860 DOI: 10.1007/s12350-020-02317-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 07/10/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND We have set out to develop a catheter-based theranostic system that: (a) identifies diseased and at-risk myocardium via endocardial detection of systemically delivered β-emitting radiotracers and (b) utilizes molecular signals to guide delivery of therapeutics to appropriate tissue via direct intramyocardial injection. METHODS Our prototype device consists of a miniature β-radiation detector contained within the tip of a flexible intravascular catheter. The catheter can be adapted to incorporate an injection port and retractable needle for therapeutic delivery. The performance of the β-detection catheter was assessed in vitro with various β-emitting radionuclides and ex vivo in hearts of pigs following systemic injection of 18F-fluorodeoxyglucose (18F-FDG) at 1-week post-myocardial infarction. Regional catheter-based endocardial measurements of 18F activity were compared to regional tissue activity from PET/CT images and gamma counting. RESULTS The β-detection catheter demonstrated sensitive in vitro detection of β-radiation from 22Na (β+), 18F (β+), and 204Tl (β-), with minimal sensitivity to γ-radiation. For 18F, the catheter demonstrated a sensitivity of 4067 counts/s/μCi in contact and a spatial resolution of 1.1 mm FWHM. Ex vivo measurements of endocardial 18F activity with the β-detection catheter in the chronic pig infarct model demonstrated good qualitative and quantitative correlation with regional tissue activity from PET/CT images and gamma counting. CONCLUSION The prototype β-detection catheter demonstrates sensitive and selective detection of β- and β+ emissions over a wide range of energies and enables high-fidelity ex vivo characterization of endocardial activity from systemically delivered 18F-FDG.
Collapse
Affiliation(s)
- John C Stendahl
- Section of Cardiovascular Medicine, Department of Medicine, Yale Translational Research Imaging Center, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Zhao Liu
- Department of Biomedical Engineering, Yale University, School of Engineering and Applied Science, New Haven, CT, 06520, USA
| | - Nabil E Boutagy
- Section of Cardiovascular Medicine, Department of Medicine, Yale Translational Research Imaging Center, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Eliahoo Nataneli
- IntraMedical Imaging, LLC, 12569 Crenshaw Blvd, Hawthorne, CA, 90250, USA
| | - Farhad Daghighian
- IntraMedical Imaging, LLC, 12569 Crenshaw Blvd, Hawthorne, CA, 90250, USA
| | - Albert J Sinusas
- Section of Cardiovascular Medicine, Department of Medicine, Yale Translational Research Imaging Center, Yale University School of Medicine, New Haven, CT, 06520, USA.
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, P.O. Box 208017, Dana 3, New Haven, CT, 06520-8017, USA.
- Department of Biomedical Engineering, Yale University, School of Engineering and Applied Science, New Haven, CT, 06520, USA.
| |
Collapse
|
27
|
Thackeray JT. Fantastic voyage: Catheter-based quantification of tracer distribution on a miniature scale. J Nucl Cardiol 2022; 29:677-679. [PMID: 33025474 PMCID: PMC8993770 DOI: 10.1007/s12350-020-02379-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 11/01/2022]
Affiliation(s)
- James T Thackeray
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg Str 1, Hannover, 30625, Germany.
| |
Collapse
|
28
|
Comparison of transient and permanent LAD ligation in mice using 18F-FDG PET imaging. Ann Nucl Med 2022; 36:533-543. [PMID: 35355159 PMCID: PMC9132804 DOI: 10.1007/s12149-022-01734-8] [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: 12/12/2021] [Accepted: 02/27/2022] [Indexed: 11/10/2022]
Abstract
Objective Animal models for myocardial injuries represent important cornerstones in cardiovascular research to monitor the pathological processes and therapeutic approaches. We investigated the association of 18F-FDG derived left ventricular metabolic volume (LVMV), defect area and cardiac function in mice after permanent or transient ligation of the left anterior descending artery (LAD). Methods Serial non-invasive ECG-gated 2-deoxy-2-[18F]fluoro-d-glucose positron emission tomography (18F-FDG PET) after permanent or transient LAD ligation enabled a longitudinal in vivo correlation of 18F-FDG derived left ventricular metabolic volume to functional parameters and myocardial defect. Results The LVMV shows a more prominent drop after permanent than transient LAD ligation and recovers after 30 days. The loss of LVMV correlates with the defect area assessed by QPS software. Cardiac function parameters (e.g., EDV, ESV, SV) by the QGS software positively correlate with LVMV after permanent and transient LAD ligation. Conclusions This study provides novel insight into 18F-FDG derived LVMV after permanent and transient LAD ligation by longitudinal in 18F-FDG PET imaging and underlines the associations of the FDG derived parameter and cardiac function. Supplementary Information The online version contains supplementary material available at 10.1007/s12149-022-01734-8.
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW Successful treatment of cancer can be hampered by the attendant risk of cardiotoxicity, manifesting as cardiomyopathy, left ventricle systolic dysfunction and, in some cases, heart failure. This risk can be mitigated if the injury to the heart is detected before the onset to irreversible cardiac impairment. The gold standard for cardiac imaging in cardio-oncology is echocardiography. Despite improvements in the application of this modality, it is not typically sensitive to sub-clinical or early-stage dysfunction. We identify in this review some emerging tracers for detecting incipient cardiotoxicity by positron emission tomography (PET). RECENT FINDINGS Vectors labeled with positron-emitting radionuclides (e.g., carbon-11, fluorine-18, gallium-68) are now available to study cardiac function, metabolism, and tissue repair in preclinical models. Many of these probes are highly sensitive to early damage, thereby potentially addressing the limitations of current imaging approaches, and show promise in preliminary clinical evaluations. The overlapping pathophysiology between cardiotoxicity and heart failure significantly expands the number of imaging tools available to cardio-oncology. This is highlighted by the emergence of radiolabeled probes targeting fibroblast activation protein (FAP) for sensitive detection of dysregulated healing process that underpins adverse cardiac remodeling. The growth of PET scanner technology also creates an opportunity for a renaissance in metabolic imaging in cardio-oncology research.
Collapse
Affiliation(s)
- James M. Kelly
- Division of Radiopharmaceutical Sciences and Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, Belfer Research Building, Room BB-1604, 413 East 69th St, New York, NY 10021 USA
- Citigroup Biomedical Imaging Center, Weill Cornell Medicine, New York, NY 10021 USA
| | - John W. Babich
- Division of Radiopharmaceutical Sciences and Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, Belfer Research Building, Room BB-1604, 413 East 69th St, New York, NY 10021 USA
- Citigroup Biomedical Imaging Center, Weill Cornell Medicine, New York, NY 10021 USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021 USA
| |
Collapse
|
30
|
Nuclear Molecular Imaging of Cardiac Remodeling after Myocardial Infarction. Pharmaceuticals (Basel) 2022; 15:ph15020183. [PMID: 35215296 PMCID: PMC8875369 DOI: 10.3390/ph15020183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 01/28/2022] [Accepted: 01/28/2022] [Indexed: 12/03/2022] Open
Abstract
The role of molecular imaging technologies in detecting, evaluating, and monitoring cardiovascular disease and their treatment is expanding rapidly. Gradually replacing the conventional anatomical or physiological approaches, molecular imaging strategies using biologically targeted markers provide unique insight into pathobiological processes at molecular and cellular levels and allow for cardiovascular disease evaluation and individualized therapy. This review paper will discuss currently available and developing molecular-based single-photon emission computed tomography (SPECT) and positron emission tomography (PET) imaging strategies to evaluate post-infarction cardiac remodeling. These approaches include potential targeted methods of evaluating critical biological processes, such as inflammation, angiogenesis, and scar formation.
Collapse
|
31
|
Bönner F, Gastl M, Nienhaus F, Rothe M, Jahn A, Pfeiler S, Gross U, Schultheiss HP, Ibanez B, Kozerke S, Szendroedi J, Roden M, Westenfeld R, Schrader J, Flögel U, Heusch G, Kelm M. Regional analysis of inflammation and contractile function in reperfused acute myocardial infarction by in vivo 19F cardiovascular magnetic resonance in pigs. Basic Res Cardiol 2022; 117:21. [PMID: 35389088 PMCID: PMC8989832 DOI: 10.1007/s00395-022-00928-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/11/2022] [Accepted: 03/22/2022] [Indexed: 01/31/2023]
Abstract
Inflammatory cell infiltration is central to healing after acute myocardial infarction (AMI). The relation of regional inflammation to edema, infarct size (IS), microvascular obstruction (MVO), intramyocardial hemorrhage (IMH), and regional and global LV function is not clear. Here we noninvasively characterized regional inflammation and contractile function in reperfused AMI in pigs using fluorine (19F) cardiovascular magnetic resonance (CMR). Adult anesthetized pigs underwent left anterior descending coronary artery instrumentation with either 90 min occlusion (n = 17) or without occlusion (sham, n = 5). After 3 days, in surviving animals a perfluorooctyl bromide nanoemulsion was infused intravenously to label monocytes/macrophages. At day 6, in vivo 1H-CMR was performed with cine, T2 and T2* weighted imaging, T2 and T1 mapping, perfusion and late gadolinium enhancement followed by 19F-CMR. Pigs were sacrificed for subsequent ex vivo scans and histology. Edema extent was 35 ± 8% and IS was 22 ± 6% of LV mass. Six of ten surviving AMI animals displayed both MVO and IMH (3.3 ± 1.6% and 1.9 ± 0.8% of LV mass). The 19F signal, reflecting the presence and density of monocytes/macrophages, was consistently smaller than edema volume or IS and not apparent in remote areas. The 19F signal-to-noise ratio (SNR) > 8 in the infarct border zone was associated with impaired remote systolic wall thickening. A whole heart value of 19F integral (19F SNR × milliliter) > 200 was related to initial LV remodeling independently of edema, IS, MVO, and IMH. Thus, 19F-CMR quantitatively characterizes regional inflammation after AMI and its relation to edema, IS, MVO, IMH and regional and global LV function and remodeling.
Collapse
Affiliation(s)
- Florian Bönner
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty Heinrich Heine University, Moorenstr. 5, 40225, Düsseldorf, Germany.
| | - M Gastl
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty Heinrich Heine University, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - F Nienhaus
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty Heinrich Heine University, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - M Rothe
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Düsseldorf, Germany
- German Center for Diabetes Research, Partner, Düsseldorf, Germany
| | - A Jahn
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty Heinrich Heine University, Moorenstr. 5, 40225, Düsseldorf, Germany
- Central Animal Research Facility, Heinrich Heine University, Düsseldorf, Germany
| | - S Pfeiler
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty Heinrich Heine University, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - U Gross
- Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, Germany
| | - H-P Schultheiss
- Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, Germany
| | - B Ibanez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- IIS-Fundación Jiménez Díaz Hospital, Madrid, Spain
- CIBERCV, Madrid, Spain
| | - S Kozerke
- Institute for Biomedical Engineering, University and ETH Zurich, Zurich, Switzerland
| | - J Szendroedi
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - M Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Düsseldorf, Germany
- German Center for Diabetes Research, Partner, Düsseldorf, Germany
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - R Westenfeld
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty Heinrich Heine University, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - J Schrader
- Department of Molecular Cardiology, Heinrich Heine University, Düsseldorf, Germany
- Experimental Cardiovascular Imaging, Department of Molecular Cardiology, Heinrich Heine University, Düsseldorf, Germany
| | - U Flögel
- Department of Molecular Cardiology, Heinrich Heine University, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - G Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany
| | - M Kelm
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty Heinrich Heine University, Moorenstr. 5, 40225, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
32
|
Hess A, Borchert T, Ross TL, Bengel FM, Thackeray JT. Characterizing the transition from immune response to tissue repair after myocardial infarction by multiparametric imaging. Basic Res Cardiol 2022; 117:14. [PMID: 35275268 PMCID: PMC8917105 DOI: 10.1007/s00395-022-00922-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/01/2022] [Accepted: 02/16/2022] [Indexed: 01/31/2023]
Abstract
Persistent inflammation following myocardial infarction (MI) precipitates adverse outcome including acute ventricular rupture and chronic heart failure. Molecular imaging allows longitudinal assessment of immune cell activity in the infarct territory and predicts severity of remodeling. We utilized a multiparametric imaging platform to assess the immune response and cardiac healing following MI in mice. Suppression of circulating macrophages prior to MI paradoxically resulted in higher total leukocyte content in the heart, demonstrated by increased CXC motif chemokine receptor 4 (CXCR4) positron emission tomography imaging. This supported the formation of a thrombus overlying the injured region, as identified by magnetic resonance imaging. The injured and thrombotic region in macrophage depeleted mice subsequently showed active calcification, as evidenced by accumulation of 18F-fluoride and by cardiac computed tomography. Importantly, macrophage suppression triggered a prolonged inflammatory response confirmed by post-mortem tissue analysis that was associated with higher mortality from ventricular rupture early after occlusion and with increased infarct size and worse chronic contractile function at 6 weeks after reperfusion. These findings establish a molecular imaging toolbox for monitoring the interplay between adverse immune response and tissue repair after MI. This may serve as a foundation for development and monitoring of novel targeted therapies that may include immune modulation and endogenous healing support.
Collapse
Affiliation(s)
- Annika Hess
- grid.10423.340000 0000 9529 9877Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Tobias Borchert
- grid.10423.340000 0000 9529 9877Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany ,Present Address: Cardior Pharmaceuticals GmbH, Hannover, Germany
| | - Tobias L. Ross
- grid.10423.340000 0000 9529 9877Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Frank M. Bengel
- grid.10423.340000 0000 9529 9877Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - James T. Thackeray
- grid.10423.340000 0000 9529 9877Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| |
Collapse
|
33
|
Li X, Yang W, Ma W, Zhou X, Quan Z, Li G, Liu D, Zhang Q, Han D, Gao B, Li C, Wang J, Kang F. 18F-FDG PET imaging-monitored anti-inflammatory therapy for acute myocardial infarction: Exploring the role of MCC950 in murine model. J Nucl Cardiol 2021; 28:2346-2357. [PMID: 32016690 DOI: 10.1007/s12350-020-02044-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 01/13/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND MCC950 is a novel NLRP3 inflammasome inhibitor that possesses potent anti-inflammatory properties in acute myocardial infarction (AMI). However, the lack of noninvasive monitoring methods limits its potential clinical translation. Thus, we sought to investigate whether 18F-FDG PET imaging can monitor the therapeutic effects of MCC950 in an AMI murine model. METHODS C57BL/6 mice were used to generate an AMI model. MCC950 or sterile saline was intraperitoneally administered 1 hour after surgery and then daily for 7 consecutive days. 18F-FDG PET (inflammation) imaging was used to monitor inflammatory changes on days 3 and 5. Immunohistochemistry and Western blot were used to detect inflammatory markers and to confirm the PET imaging results. 18F-FDG PET (viability) imaging was used to quantitate the viability defect expansion on days 7 and 28. Cardiac ultrasound and survival analyses were performed to evaluate the cardiac function and survival rate. Adverse remodeling was determined by Wheat Germ Agglutinin (WGA) and Masson trichrome staining. RESULTS The FDG-PET (inflammation) imaging revealed that MCC950 treatment led to lower 18F-FDG inflammatory uptakes, at the infarct region, on days 3 and 5 when compared to the MI group. The decreased M1 macrophages and neutrophils infiltration and the remission of the NLRP3/IL-1β pathway, confirmed the FDG-PET (inflammation) imaging results. The FDG-PET (viability) imaging revealed that MCC950 significantly decreased the expansion of the viability defect, demonstrating its myocardial preservation effects. The acute FDG-PET (inflammation) signal positively correlated with the late viability defect and with the reduction in the left ventricular ejection fraction (LVEF). Additionally, the alleviated adverse remodeling and the improved survival rate further support the anti-inflammatory efficiency of MCC950 in AMI. CONCLUSION Using 18F-FDG PET imaging, we noninvasively demonstrated the therapeutic effects of MCC950 in AMI and showed that 18F-FDG PET imaging holds promising application potentials in MCC950's clinical translation.
Collapse
Affiliation(s)
- Xiang Li
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Weidong Yang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Wenhui Ma
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiang Zhou
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhiyong Quan
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Guoquan Li
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Daliang Liu
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Qingju Zhang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Dong Han
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Beilei Gao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 210032, China
| | - Congye Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Jing Wang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| | - Fei Kang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
34
|
Balogh V, MacAskill MG, Hadoke PWF, Gray GA, Tavares AAS. Positron Emission Tomography Techniques to Measure Active Inflammation, Fibrosis and Angiogenesis: Potential for Non-invasive Imaging of Hypertensive Heart Failure. Front Cardiovasc Med 2021; 8:719031. [PMID: 34485416 PMCID: PMC8416043 DOI: 10.3389/fcvm.2021.719031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/22/2021] [Indexed: 12/11/2022] Open
Abstract
Heart failure, which is responsible for a high number of deaths worldwide, can develop due to chronic hypertension. Heart failure can involve and progress through several different pathways, including: fibrosis, inflammation, and angiogenesis. Early and specific detection of changes in the myocardium during the transition to heart failure can be made via the use of molecular imaging techniques, including positron emission tomography (PET). Traditional cardiovascular PET techniques, such as myocardial perfusion imaging and sympathetic innervation imaging, have been established at the clinical level but are often lacking in pathway and target specificity that is important for assessment of heart failure. Therefore, there is a need to identify new PET imaging markers of inflammation, fibrosis and angiogenesis that could aid diagnosis, staging and treatment of hypertensive heart failure. This review will provide an overview of key mechanisms underlying hypertensive heart failure and will present the latest developments in PET probes for detection of cardiovascular inflammation, fibrosis and angiogenesis. Currently, selective PET probes for detection of angiogenesis remain elusive but promising PET probes for specific targeting of inflammation and fibrosis are rapidly progressing into clinical use.
Collapse
Affiliation(s)
- Viktoria Balogh
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Imaging, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Mark G. MacAskill
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Imaging, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Patrick W. F. Hadoke
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Gillian A. Gray
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Adriana A. S. Tavares
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Imaging, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
35
|
Borchert T, Beitar L, Langer LBN, Polyak A, Wester HJ, Ross TL, Hilfiker-Kleiner D, Bengel FM, Thackeray JT. Dissecting the target leukocyte subpopulations of clinically relevant inflammation radiopharmaceuticals. J Nucl Cardiol 2021; 28:1636-1645. [PMID: 31659697 DOI: 10.1007/s12350-019-01929-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/29/2019] [Accepted: 10/01/2019] [Indexed: 10/25/2022]
Abstract
BACKGROUND Leukocyte subtypes bear distinct pro-inflammatory, reparative, and regulatory functions. Imaging inflammation provides information on disease prognosis and may guide therapy, but the cellular basis of the signal remains equivocal. We evaluated leukocyte subtype specificity of characterized clinically relevant inflammation-targeted radiotracers. METHODS AND RESULTS Leukocyte populations were purified from blood- and THP-1-derived macrophages were polarized into M1-, reparative M2a-, or M2c-macrophages. In vitro uptake assays were conducted using tracers of enhanced glucose or amino acid metabolism and molecular markers of inflammatory cells. Both 18F-deoxyglucose (18F-FDG) and the labeled amino acid 11C-methionine (11C-MET) displayed higher uptake in neutrophils and monocytes compared to other leukocytes (P = 0.005), and markedly higher accumulation in pro-inflammatory M1-macrophages compared to reparative M2a-macrophages (P < 0.001). Molecular tracers 68Ga-DOTATATE targeting the somatostatin receptor type 2 and 68Ga-pentixafor targeting the chemokine receptor type 4 (CXCR4) exhibited broad uptake by leukocyte subpopulations and polarized macrophages with highest uptake in T-cells/natural killer cells and B-cells compared to neutrophils. Mitochondrial translocator protein (TSPO)-targeted 18F-flutriciclamide selectively accumulated in monocytes and pro-inflammatory M1 macrophages (P < 0.001). Uptake by myocytes and fibroblasts tended to be higher for metabolic radiotracers. CONCLUSIONS The different in vitro cellular uptake profiles may allow isolation of distinct phases of the inflammatory pathway with specific inflammation-targeted radiotracers. The pathogenetic cell population in specific inflammatory diseases should be considered in the selection of an appropriate imaging agent.
Collapse
Affiliation(s)
- Tobias Borchert
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, 30625, Hannover, Germany
| | - Laura Beitar
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, 30625, Hannover, Germany
| | - Laura B N Langer
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, 30625, Hannover, Germany
| | - Andras Polyak
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, 30625, Hannover, Germany
| | - Hans-Jürgen Wester
- Department of Radiopharmaceutical Chemistry, Technical University of Munich, Munich, Germany
| | - Tobias L Ross
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, 30625, Hannover, Germany
| | | | - Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, 30625, Hannover, Germany
| | - James T Thackeray
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW To examine the use of positron emission tomography (PET) for imaging post-infarct myocardial inflammation and repair. RECENT FINDINGS Dysregulated immune responses after myocardial infarction are associated with adverse cardiac remodelling and an increased likelihood of ischaemic heart failure. PET imaging utilising novel tracers can be applied to visualise different components of the post-infarction inflammatory and repair processes. This approach could offer unique pathophysiological insights that could prove useful for the identification and risk-stratification of individuals who would ultimately benefit most from emerging immune-modulating therapies. PET imaging could also bridge the clinical translational gap as a surrogate measure of drug efficacy in early-stage clinical trials in patients with myocardial infarction. The use of hybrid PET/MR imaging, in particular, offers the additional advantage of simultaneous in vivo molecular imaging and detailed assessment of myocardial function, viability and tissue characterisation. Further research is needed to realise the true clinical translational value of PET imaging after myocardial infarction.
Collapse
Affiliation(s)
- Andrej Ćorović
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Meritxell Nus
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Ziad Mallat
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - James H. F. Rudd
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Jason M. Tarkin
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
37
|
Glasenapp A, Derlin K, Gutberlet M, Hess A, Ross TL, Wester HJ, Bengel FM, Thackeray JT. Molecular Imaging of Inflammation and Fibrosis in Pressure Overload Heart Failure. Circ Res 2021; 129:369-382. [PMID: 34074134 DOI: 10.1161/circresaha.120.318539] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Aylina Glasenapp
- Department of Nuclear Medicine (A.G., A.H., T.L.R., F.M.B., J.T.T.)
- Institute of Diagnostic and Interventional Radiology, Hannover Medical School, Germany (A.G., K.D., M.G.)
| | - Katja Derlin
- Institute of Diagnostic and Interventional Radiology, Hannover Medical School, Germany (A.G., K.D., M.G.)
| | - Marcel Gutberlet
- Institute of Diagnostic and Interventional Radiology, Hannover Medical School, Germany (A.G., K.D., M.G.)
| | - Annika Hess
- Department of Nuclear Medicine (A.G., A.H., T.L.R., F.M.B., J.T.T.)
| | - Tobias L Ross
- Department of Nuclear Medicine (A.G., A.H., T.L.R., F.M.B., J.T.T.)
| | - Hans-Jürgen Wester
- Technical University of Munich, Radiopharmaceutical Chemistry, Germany (H.-J.W.)
| | - Frank M Bengel
- Department of Nuclear Medicine (A.G., A.H., T.L.R., F.M.B., J.T.T.)
| | | |
Collapse
|
38
|
Hess A, Derlin T, Koenig T, Diekmann J, Wittneben A, Wang Y, Wester HJ, Ross TL, Wollert KC, Bauersachs J, Bengel FM, Thackeray JT. Molecular imaging-guided repair after acute myocardial infarction by targeting the chemokine receptor CXCR4. Eur Heart J 2021; 41:3564-3575. [PMID: 32901270 DOI: 10.1093/eurheartj/ehaa598] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/27/2020] [Accepted: 07/02/2020] [Indexed: 01/04/2023] Open
Abstract
AIMS Balance between inflammatory and reparative leucocytes allows optimal healing after myocardial infarction (MI). Interindividual heterogeneity evokes variable functional outcome complicating targeted therapy. We aimed to characterize infarct chemokine CXC-motif receptor 4 (CXCR4) expression using positron emission tomography (PET) and establish its relationship to cardiac outcome. We tested whether image-guided early CXCR4 directed therapy attenuates chronic dysfunction. METHODS AND RESULTS Mice (n = 180) underwent coronary ligation or sham surgery and serial PET imaging over 7 days. Infarct CXCR4 content was elevated over 3 days after MI compared with sham (%ID/g, Day 1:1.1 ± 0.2; Day 3:0.9 ± 0.2 vs. 0.6 ± 0.1, P < 0.001), confirmed by flow cytometry and histopathology. Mice that died of left ventricle (LV) rupture exhibited persistent inflammation at 3 days compared with survivors (1.2 ± 0.3 vs. 0.9 ± 0.2% ID/g, P < 0.001). Cardiac magnetic resonance measured cardiac function. Higher CXCR4 signal at 1 and 3 days independently predicted worse functional outcome at 6 weeks (rpartial = -0.4, P = 0.04). Mice were treated with CXCR4 blocker AMD3100 following the imaging timecourse. On-peak CXCR4 blockade at 3 days lowered LV rupture incidence vs. untreated MI (8% vs. 25%), and improved contractile function at 6 weeks (+24%, P = 0.01). Off-peak CXCR4 blockade at 7 days did not improve outcome. Flow cytometry analysis revealed lower LV neutrophil and Ly6Chigh monocyte content after on-peak treatment. Patients (n = 50) early after MI underwent CXCR4 PET imaging and functional assessment. Infarct CXCR4 expression in acute MI patients correlated with contractile function at time of PET and on follow-up. CONCLUSION Positron emission tomography imaging identifies early CXCR4 up-regulation which predicts acute rupture and chronic contractile dysfunction. Imaging-guided CXCR4 inhibition accelerates inflammatory resolution and improves outcome. This supports a molecular imaging-based theranostic approach to guide therapy after MI.
Collapse
Affiliation(s)
- Annika Hess
- Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Tobias Koenig
- D epartment of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Johanna Diekmann
- Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Alexander Wittneben
- Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Yong Wang
- D epartment of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Hans-Juergen Wester
- Radiopharmaceutical Chemistry, Technical University of Munich, Walther-Meissner-Str. 3, 85748 Garching, Germany
| | - Tobias L Ross
- Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Kai C Wollert
- D epartment of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Johann Bauersachs
- D epartment of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - James T Thackeray
- Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| |
Collapse
|
39
|
Wahl RL, Chareonthaitawee P, Clarke B, Drzezga A, Lindenberg L, Rahmim A, Thackeray J, Ulaner GA, Weber W, Zukotynski K, Sunderland J. Mars Shot for Nuclear Medicine, Molecular Imaging, and Molecularly Targeted Radiopharmaceutical Therapy. J Nucl Med 2021; 62:6-14. [PMID: 33334911 DOI: 10.2967/jnumed.120.253450] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/23/2020] [Indexed: 02/06/2023] Open
Abstract
The Society of Nuclear Medicine and Molecular Imaging created the Value Initiative in 2017 as a major component of its strategic plan to further demonstrate the value of molecular imaging and molecularly targeted radiopharmaceutical therapy to patients, physicians, payers, and funding agencies. The research and discovery domain, 1 of 5 under the Value Initiative, has a goal of advancing the research and development of diagnostic and therapeutic nuclear medicine. Research and discovery efforts and achievements are essential to ensure a bright future for NM and to translate science to practice. Given the remarkable progress in the field, leaders from the research and discovery domain and society councils identified 5 broad areas of opportunity with potential for substantive growth and clinical impact. This article discusses these 5 growth areas, identifying specific areas of particularly high importance for future study and development. As there was an understanding that goals should be both visionary yet achievable, this effort was called the Mars shot for nuclear medicine.
Collapse
Affiliation(s)
- Richard L Wahl
- Mallinckrodt Institute of Radiology, Washington University St. Louis, Missouri
| | | | - Bonnie Clarke
- Research and Discovery, Society of Nuclear Medicine and Molecular Imaging, Reston, Virginia
| | - Alexander Drzezga
- Department of Nuclear Medicine, University of Cologne, Cologne, Germany, German Center for Neurodegenerative Diseases, Bonn-Cologne, Germany, and Institute of Neuroscience and Medicine, Molecular Organization of the Brain, Forschungszentrum Jülich, Jülich, Germany
| | - Liza Lindenberg
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Arman Rahmim
- Departments of Radiology and Physics, University of British Columbia, Vancouver, British Columbia, Canada; Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - James Thackeray
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Gary A Ulaner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, and Molecular Imaging and Therapy, Hoag Cancer Center, Newport Beach, California
| | - Wolfgang Weber
- Department of Nuclear Medicine, Technical University Munich, Munich, Germany
| | - Katherine Zukotynski
- Departments of Medicine and Radiology, McMaster University, Hamilton, Ontario, Canada; and
| | - John Sunderland
- Departments of Radiology and Physics, University of Iowa, Iowa City, Iowa
| |
Collapse
|
40
|
Imaging Inflammation with Positron Emission Tomography. Biomedicines 2021; 9:biomedicines9020212. [PMID: 33669804 PMCID: PMC7922638 DOI: 10.3390/biomedicines9020212] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 01/28/2021] [Accepted: 02/12/2021] [Indexed: 12/19/2022] Open
Abstract
The impact of inflammation on the outcome of many medical conditions such as cardiovascular diseases, neurological disorders, infections, cancer, and autoimmune diseases has been widely acknowledged. However, in contrast to neurological, oncologic, and cardiovascular disorders, imaging plays a minor role in research and management of inflammation. Imaging can provide insights into individual and temporospatial biology and grade of inflammation which can be of diagnostic, therapeutic, and prognostic value. There is therefore an urgent need to evaluate and understand current approaches and potential applications for imaging of inflammation. This review discusses radiotracers for positron emission tomography (PET) that have been used to image inflammation in cardiovascular diseases and other inflammatory conditions with a special emphasis on radiotracers that have already been successfully applied in clinical settings.
Collapse
|
41
|
Congestive Heart Failure. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00050-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
42
|
Hess A, Thackeray JT, Wollert KC, Bengel FM. Radionuclide Image-Guided Repair of the Heart. JACC Cardiovasc Imaging 2020; 13:2415-2429. [DOI: 10.1016/j.jcmg.2019.11.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/23/2019] [Accepted: 11/05/2019] [Indexed: 01/12/2023]
|
43
|
Nahrendorf M, Hoyer FF, Meerwaldt AE, van Leent MM, Senders ML, Calcagno C, Robson PM, Soultanidis G, Pérez-Medina C, Teunissen AJ, Toner YC, Ishikawa K, Fish K, Sakurai K, van Leeuwen EM, Klein ED, Sofias AM, Reiner T, Rohde D, Aguirre AD, Wojtkiewicz G, Schmidt S, Iwamoto Y, Izquierdo-Garcia D, Caravan P, Swirski FK, Weissleder R, Mulder WJ. Imaging Cardiovascular and Lung Macrophages With the Positron Emission Tomography Sensor 64Cu-Macrin in Mice, Rabbits, and Pigs. Circ Cardiovasc Imaging 2020; 13:e010586. [PMID: 33076700 PMCID: PMC7583675 DOI: 10.1161/circimaging.120.010586] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/28/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Macrophages, innate immune cells that reside in all organs, defend the host against infection and injury. In the heart and vasculature, inflammatory macrophages also enhance tissue damage and propel cardiovascular diseases. METHODS We here use in vivo positron emission tomography (PET) imaging, flow cytometry, and confocal microscopy to evaluate quantitative noninvasive assessment of cardiac, arterial, and pulmonary macrophages using the nanotracer 64Cu-Macrin-a 20-nm spherical dextran nanoparticle assembled from nontoxic polyglucose. RESULTS PET imaging using 64Cu-Macrin faithfully reported accumulation of macrophages in the heart and lung of mice with myocardial infarction, sepsis, or pneumonia. Flow cytometry and confocal microscopy detected the near-infrared fluorescent version of the nanoparticle (VT680Macrin) primarily in tissue macrophages. In 5-day-old mice, 64Cu-Macrin PET imaging quantified physiologically more numerous cardiac macrophages. Upon intravenous administration of 64Cu-Macrin in rabbits and pigs, we detected heightened macrophage numbers in the infarcted myocardium, inflamed lung regions, and atherosclerotic plaques using a clinical PET/magnetic resonance imaging scanner. Toxicity studies in rats and human dosimetry estimates suggest that 64Cu-Macrin is safe for use in humans. CONCLUSIONS Taken together, these results indicate 64Cu-Macrin could serve as a facile PET nanotracer to survey spatiotemporal macrophage dynamics during various physiological and pathological conditions. 64Cu-Macrin PET imaging could stage inflammatory cardiovascular disease activity, assist disease management, and serve as an imaging biomarker for emerging macrophage-targeted therapeutics.
Collapse
Affiliation(s)
- Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Internal Medicine I, University Hospital Wuerzburg, Wuerzburg, Germany
- These authors contributed equally: Matthias Nahrendorf, Friedrich Felix Hoyer
| | - Friedrich Felix Hoyer
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- These authors contributed equally: Matthias Nahrendorf, Friedrich Felix Hoyer
| | - Anu E. Meerwaldt
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands
| | - Mandy M.T. van Leent
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medical Biochemistry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Max L. Senders
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medical Biochemistry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Claudia Calcagno
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Philip M. Robson
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - George Soultanidis
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carlos Pérez-Medina
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Abraham J.P. Teunissen
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yohana C. Toner
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kiyotake Ishikawa
- Cardiovascular Research Center, Department of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kenneth Fish
- Cardiovascular Research Center, Department of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ken Sakurai
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Esther M. van Leeuwen
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands
| | - Emma D. Klein
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexandros Marios Sofias
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Thomas Reiner
- Department of Radiology and Chemical Biology Program, Memorial Sloan-Kettering Cancer Center; Department of Radiology, Weill Cornell Medical College, New York, NY, USA
| | - David Rohde
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Aaron D. Aguirre
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Gregory Wojtkiewicz
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Stephen Schmidt
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Yoshiko Iwamoto
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - David Izquierdo-Garcia
- The Institute for Innovation in Imaging, A. A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Peter Caravan
- The Institute for Innovation in Imaging, A. A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Filip K. Swirski
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- The Institute for Innovation in Imaging, A. A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Willem J.M. Mulder
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medical Biochemistry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
44
|
Abstract
Cardiac PET/MR imaging is an integrated imaging approach that requires less radiation than PET/computed tomography and combines the high spatial resolution and morphologic data from MR imaging with the physiologic information from PET. This hybrid approach has the potential to improve the diagnostic and prognostic evaluation of several cardiovascular conditions, such as ischemic heart disease, infiltrative diseases such as sarcoidosis, acute and chronic myocarditis, and cardiac masses. Herein, the authors discuss the strengths of PET and MR imaging in several cardiovascular conditions; the challenges and potential; and the current data on the application of this powerful hybrid imaging modality.
Collapse
Affiliation(s)
- Rhanderson Cardoso
- Division of Cardiology, Johns Hopkins Hospital, 600 North Wolfe Street, Blalock 547, Baltimore, MD 21287, USA
| | - Thorsten M Leucker
- Division of Cardiology, Johns Hopkins Hospital, 600 North Wolfe Street, Blalock 547, Baltimore, MD 21287, USA.
| |
Collapse
|
45
|
Haider A, Bengs S, Schade K, Wijnen WJ, Portmann A, Etter D, Fröhlich S, Warnock GI, Treyer V, Burger IA, Fiechter M, Kudura K, Fuchs TA, Pazhenkottil AP, Buechel RR, Kaufmann PA, Meisel A, Stolzmann P, Gebhard C. Myocardial 18F-FDG Uptake Pattern for Cardiovascular Risk Stratification in Patients Undergoing Oncologic PET/CT. J Clin Med 2020; 9:E2279. [PMID: 32709049 PMCID: PMC7408629 DOI: 10.3390/jcm9072279] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/11/2020] [Accepted: 07/14/2020] [Indexed: 12/03/2022] Open
Abstract
OBJECTIVE Positron emission tomography/computed tomography with 18F-fluorodeoxy-glucose (18F-FDG-PET/CT) has become the standard staging modality in various tumor entities. Cancer patients frequently receive cardio-toxic therapies. However, routine cardiovascular assessment in oncologic patients is not performed in current clinical practice. Accordingly, this study sought to assess whether myocardial 18F-FDG uptake patterns of patients undergoing oncologic PET/CT can be used for cardiovascular risk stratification. METHODS Myocardial 18F-FDG uptake pattern was assessed in 302 patients undergoing both oncologic whole-body 18F-FDG-PET/CT and myocardial perfusion imaging by single-photon emission computed tomography (SPECT-MPI) within a six-month period. Primary outcomes were myocardial 18F-FDG uptake pattern, impaired myocardial perfusion, ongoing ischemia, myocardial scar, and left ventricular ejection fraction. RESULTS Among all patients, 109 (36.1%) displayed no myocardial 18F-FDG uptake, 77 (25.5%) showed diffuse myocardial 18F-FDG uptake, 24 (7.9%) showed focal 18F-FDG uptake, and 92 (30.5%) had a focal on diffuse myocardial 18F-FDG uptake pattern. In contrast to the other uptake patterns, focal myocardial 18F-FDG uptake was predominantly observed in patients with myocardial abnormalities (i.e., abnormal perfusion, impaired LVEF, myocardial ischemia, or scar). Accordingly, a multivariate logistic regression identified focal myocardial 18F-FDG uptake as a strong predictor of abnormal myocardial function/perfusion (odds ratio (OR) 5.32, 95% confidence interval (CI) 1.73-16.34, p = 0.003). Similarly, focal myocardial 18F-FDG uptake was an independent predictor of ongoing ischemia and myocardial scar (OR 4.17, 95% CI 1.53-11.4, p = 0.005 and OR 3.78, 95% CI 1.47-9.69, p = 0.006, respectively). CONCLUSIONS Focal myocardial 18F-FDG uptake seen on oncologic PET/CT indicates a significantly increased risk for multiple myocardial abnormalities. Obtaining and taking this information into account will help to stratify patients according to risk and will reduce unnecessary cardiovascular complications in cancer patients.
Collapse
Affiliation(s)
- Ahmed Haider
- Department of Nuclear Medicine, University Hospital Zurich, 8091 Zurich, Switzerland; (S.B.); (K.S.); (W.J.W.); (A.P.); (D.E.); (S.F.); (G.I.W.); (V.T.); (I.A.B.); (M.F.); (K.K.); (T.A.F.); (A.P.P.); (R.R.B.); (P.A.K.); (A.M.); (P.S.); (C.G.)
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland
| | - Susan Bengs
- Department of Nuclear Medicine, University Hospital Zurich, 8091 Zurich, Switzerland; (S.B.); (K.S.); (W.J.W.); (A.P.); (D.E.); (S.F.); (G.I.W.); (V.T.); (I.A.B.); (M.F.); (K.K.); (T.A.F.); (A.P.P.); (R.R.B.); (P.A.K.); (A.M.); (P.S.); (C.G.)
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland
| | - Katharina Schade
- Department of Nuclear Medicine, University Hospital Zurich, 8091 Zurich, Switzerland; (S.B.); (K.S.); (W.J.W.); (A.P.); (D.E.); (S.F.); (G.I.W.); (V.T.); (I.A.B.); (M.F.); (K.K.); (T.A.F.); (A.P.P.); (R.R.B.); (P.A.K.); (A.M.); (P.S.); (C.G.)
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland
| | - Winandus J. Wijnen
- Department of Nuclear Medicine, University Hospital Zurich, 8091 Zurich, Switzerland; (S.B.); (K.S.); (W.J.W.); (A.P.); (D.E.); (S.F.); (G.I.W.); (V.T.); (I.A.B.); (M.F.); (K.K.); (T.A.F.); (A.P.P.); (R.R.B.); (P.A.K.); (A.M.); (P.S.); (C.G.)
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland
| | - Angela Portmann
- Department of Nuclear Medicine, University Hospital Zurich, 8091 Zurich, Switzerland; (S.B.); (K.S.); (W.J.W.); (A.P.); (D.E.); (S.F.); (G.I.W.); (V.T.); (I.A.B.); (M.F.); (K.K.); (T.A.F.); (A.P.P.); (R.R.B.); (P.A.K.); (A.M.); (P.S.); (C.G.)
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland
| | - Dominik Etter
- Department of Nuclear Medicine, University Hospital Zurich, 8091 Zurich, Switzerland; (S.B.); (K.S.); (W.J.W.); (A.P.); (D.E.); (S.F.); (G.I.W.); (V.T.); (I.A.B.); (M.F.); (K.K.); (T.A.F.); (A.P.P.); (R.R.B.); (P.A.K.); (A.M.); (P.S.); (C.G.)
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland
| | - Sandro Fröhlich
- Department of Nuclear Medicine, University Hospital Zurich, 8091 Zurich, Switzerland; (S.B.); (K.S.); (W.J.W.); (A.P.); (D.E.); (S.F.); (G.I.W.); (V.T.); (I.A.B.); (M.F.); (K.K.); (T.A.F.); (A.P.P.); (R.R.B.); (P.A.K.); (A.M.); (P.S.); (C.G.)
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland
| | - Geoffrey I. Warnock
- Department of Nuclear Medicine, University Hospital Zurich, 8091 Zurich, Switzerland; (S.B.); (K.S.); (W.J.W.); (A.P.); (D.E.); (S.F.); (G.I.W.); (V.T.); (I.A.B.); (M.F.); (K.K.); (T.A.F.); (A.P.P.); (R.R.B.); (P.A.K.); (A.M.); (P.S.); (C.G.)
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland
| | - Valerie Treyer
- Department of Nuclear Medicine, University Hospital Zurich, 8091 Zurich, Switzerland; (S.B.); (K.S.); (W.J.W.); (A.P.); (D.E.); (S.F.); (G.I.W.); (V.T.); (I.A.B.); (M.F.); (K.K.); (T.A.F.); (A.P.P.); (R.R.B.); (P.A.K.); (A.M.); (P.S.); (C.G.)
| | - Irene A. Burger
- Department of Nuclear Medicine, University Hospital Zurich, 8091 Zurich, Switzerland; (S.B.); (K.S.); (W.J.W.); (A.P.); (D.E.); (S.F.); (G.I.W.); (V.T.); (I.A.B.); (M.F.); (K.K.); (T.A.F.); (A.P.P.); (R.R.B.); (P.A.K.); (A.M.); (P.S.); (C.G.)
| | - Michael Fiechter
- Department of Nuclear Medicine, University Hospital Zurich, 8091 Zurich, Switzerland; (S.B.); (K.S.); (W.J.W.); (A.P.); (D.E.); (S.F.); (G.I.W.); (V.T.); (I.A.B.); (M.F.); (K.K.); (T.A.F.); (A.P.P.); (R.R.B.); (P.A.K.); (A.M.); (P.S.); (C.G.)
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland
- Swiss Paraplegic Center, 6207 Nottwil, Switzerland
| | - Ken Kudura
- Department of Nuclear Medicine, University Hospital Zurich, 8091 Zurich, Switzerland; (S.B.); (K.S.); (W.J.W.); (A.P.); (D.E.); (S.F.); (G.I.W.); (V.T.); (I.A.B.); (M.F.); (K.K.); (T.A.F.); (A.P.P.); (R.R.B.); (P.A.K.); (A.M.); (P.S.); (C.G.)
| | - Tobias A. Fuchs
- Department of Nuclear Medicine, University Hospital Zurich, 8091 Zurich, Switzerland; (S.B.); (K.S.); (W.J.W.); (A.P.); (D.E.); (S.F.); (G.I.W.); (V.T.); (I.A.B.); (M.F.); (K.K.); (T.A.F.); (A.P.P.); (R.R.B.); (P.A.K.); (A.M.); (P.S.); (C.G.)
| | - Aju P. Pazhenkottil
- Department of Nuclear Medicine, University Hospital Zurich, 8091 Zurich, Switzerland; (S.B.); (K.S.); (W.J.W.); (A.P.); (D.E.); (S.F.); (G.I.W.); (V.T.); (I.A.B.); (M.F.); (K.K.); (T.A.F.); (A.P.P.); (R.R.B.); (P.A.K.); (A.M.); (P.S.); (C.G.)
| | - Ronny R. Buechel
- Department of Nuclear Medicine, University Hospital Zurich, 8091 Zurich, Switzerland; (S.B.); (K.S.); (W.J.W.); (A.P.); (D.E.); (S.F.); (G.I.W.); (V.T.); (I.A.B.); (M.F.); (K.K.); (T.A.F.); (A.P.P.); (R.R.B.); (P.A.K.); (A.M.); (P.S.); (C.G.)
| | - Philipp A. Kaufmann
- Department of Nuclear Medicine, University Hospital Zurich, 8091 Zurich, Switzerland; (S.B.); (K.S.); (W.J.W.); (A.P.); (D.E.); (S.F.); (G.I.W.); (V.T.); (I.A.B.); (M.F.); (K.K.); (T.A.F.); (A.P.P.); (R.R.B.); (P.A.K.); (A.M.); (P.S.); (C.G.)
| | - Alexander Meisel
- Department of Nuclear Medicine, University Hospital Zurich, 8091 Zurich, Switzerland; (S.B.); (K.S.); (W.J.W.); (A.P.); (D.E.); (S.F.); (G.I.W.); (V.T.); (I.A.B.); (M.F.); (K.K.); (T.A.F.); (A.P.P.); (R.R.B.); (P.A.K.); (A.M.); (P.S.); (C.G.)
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland
| | - Paul Stolzmann
- Department of Nuclear Medicine, University Hospital Zurich, 8091 Zurich, Switzerland; (S.B.); (K.S.); (W.J.W.); (A.P.); (D.E.); (S.F.); (G.I.W.); (V.T.); (I.A.B.); (M.F.); (K.K.); (T.A.F.); (A.P.P.); (R.R.B.); (P.A.K.); (A.M.); (P.S.); (C.G.)
| | - Catherine Gebhard
- Department of Nuclear Medicine, University Hospital Zurich, 8091 Zurich, Switzerland; (S.B.); (K.S.); (W.J.W.); (A.P.); (D.E.); (S.F.); (G.I.W.); (V.T.); (I.A.B.); (M.F.); (K.K.); (T.A.F.); (A.P.P.); (R.R.B.); (P.A.K.); (A.M.); (P.S.); (C.G.)
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
46
|
Tarkin JM, Ćorović A, Wall C, Gopalan D, Rudd JH. Positron emission tomography imaging in cardiovascular disease. Heart 2020; 106:1712-1718. [PMID: 32571959 DOI: 10.1136/heartjnl-2019-315183] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/22/2020] [Accepted: 05/26/2020] [Indexed: 01/05/2023] Open
Abstract
Positron emission tomography (PET) imaging is useful in cardiovascular disease across several areas, from assessment of myocardial perfusion and viability, to highlighting atherosclerotic plaque activity and measuring the extent of cardiac innervation in heart failure. Other important roles of PET have emerged in prosthetic valve endocarditis, implanted device infection, infiltrative cardiomyopathies, aortic stenosis and cardio-oncology. Advances in scanner technology, including hybrid PET/MRI and total body PET imaging, as well as the development of novel PET tracers and cardiac-specific postprocessing techniques using artificial intelligence will undoubtedly continue to progress the field.
Collapse
Affiliation(s)
- Jason M Tarkin
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Andrej Ćorović
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Christopher Wall
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Deepa Gopalan
- Radiology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, Cambridgeshire, UK
| | - James Hf Rudd
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
47
|
Schwaiger M, Nekolla SG. What did we learn from PET/MR? J Nucl Cardiol 2020; 27:899-902. [PMID: 31385222 DOI: 10.1007/s12350-019-01815-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 05/23/2019] [Indexed: 11/26/2022]
Affiliation(s)
| | - Stephan G Nekolla
- Department of Nuclear Medicine, Technical University of Munich, Munich, Germany
- DZHK (Deutsches, Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
48
|
Clinical and Research Tools for the Study of Cardiovascular Effects of Cancer Therapy. J Cardiovasc Transl Res 2020; 13:417-430. [PMID: 32472498 DOI: 10.1007/s12265-020-10030-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 05/12/2020] [Indexed: 12/16/2022]
Abstract
The expansion of cancer therapeutics has paved the way for improved cancer-related outcomes. Cardiotoxicity from cancer therapy occurs in a small but significant subset of patients, is often poorly understood, and contributes to adverse outcomes at all stages of cancer treatment. Given the often-idiopathic occurrence of cardiotoxicity, novel strategies are needed for risk-stratification and early identification of cancer patients experiencing cardiotoxicity. Clinical and research tools extending from imaging to blood-based biomarkers and pluripotent stem cells are being explored as methods to study the cardiovascular impact of various cancer treatments. Here we provide an overview of tools currently available for evaluation of cardiotoxicity and highlight novel techniques in development aimed at understanding underlying pathophysiologic mechanisms.
Collapse
|
49
|
18F-FDG PET-Based Imaging of Myocardial Inflammation Following Acute Myocardial Infarction in a Mouse Model. Int J Mol Sci 2020; 21:ijms21093340. [PMID: 32397287 PMCID: PMC7246846 DOI: 10.3390/ijms21093340] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 01/13/2023] Open
Abstract
Cellular inflammation is an integral part of the healing process following acute myocardial infarction and has been under intense investigation for both therapeutic and prognostic approaches. Monocytes and macrophages are metabolically highly active and show increased uptake rates of glucose and its analog, 18F-FDG. Yet, the specific allocation of the radioactivity to the inflammatory cells via positron emission tomography (PET) imaging requires the suppression of glucose metabolism in viable myocardium. In mice, the most important model organism in basic research, this can be achieved by the application of ketamine/xylazine (KX) for anesthesia instead of isoflurane. Yet, while the consensus exists that glucose metabolism is effectively suppressed, a strategy for reproducible image analysis is grossly lacking and causes uncertainty concerning data interpretation. We introduce a simple strategy for systematic image analysis, which is a prerequisite to evaluate therapies targeting myocardial inflammation. Mice underwent permanent occlusion of the left anterior descending artery (LAD), inducing an acute myocardial infarction (MI). Five days after MI induction, 10MBq 18F-FDG was injected intravenously and a static PET/CT scan under ketamine/xylazine anesthesia was performed. For image reconstruction, we used an algorithm based on three-dimensional ordered subsets expectation maximization (3D-OSEM) followed by three-dimensional ordinary Poisson maximum a priori (MAP) reconstruction. Using this approach, high focal tracer uptake was typically located in the border zone of the infarct by visual inspection. To precisely demarcate the border zone for reproducible volume of interest (VOI) positioning, our protocol relies on positioning VOIs around the whole left ventricle, the inferobasal wall and the anterolateral wall guided by anatomical landmarks. This strategy enables comparable data in mouse studies, which is an important prerequisite for using a PET-based assessment of myocardial inflammation as a prognostic tool in therapeutic applications.
Collapse
|
50
|
Bilani N, Elson L, Martinez F, Sadler D, Nahleh Z, Elimimian E, Alley E. A Multimodal Approach to Evaluate for Cardiac Metastasis in a Case of Non-Small Cell Lung Cancer. Case Rep Oncol 2020; 13:212-218. [PMID: 32308579 PMCID: PMC7154231 DOI: 10.1159/000505534] [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: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 11/19/2022] Open
Abstract
Malignancies have demonstrated the ability to metastasize to cardiac tissue. However, an optimal diagnostic algorithm for cardiac tumors has not yet been established, due at least in part to the scarcity of symptomatic cases. Several case reports describe how the usage of <sup>18</sup>F-labeled fluorodeoxyglucose positron emission tomography (<sup>18</sup>F-FDG PET) incidentally revealed cardiac neoplasia. This modality, which indicates uptake sites of the radioisotope <sup>18</sup>F-FDG, allows for whole-body imaging and is often used for preoperative determination of malignant metastasis or for assessing response to therapy over time. However, findings of false positivity are often reported due to increased FDG avidity caused by a range of other, nonmetastatic processes, most notably inflammation and infection. In this case report, an 84-year-old male with stage IV non-small cell lung cancer presented a clinical course, echocardiogram, and <sup>18</sup>F-FDG PET-CT findings that were suggestive of endocardial metastasis. Nine months into therapy, after extensive consultation, the patient finally consented to a more complete workup using cardiac MRI (CMRI), which showed no evidence of cardiac metastasis. This case report supports the utility of CMRI as a means of further interpreting intracardiac, localized FDG uptake foci in PET-CT findings, in order to avoid false positivity and further refine proposed cardiac differential diagnoses in cancer patients.
Collapse
Affiliation(s)
- Nadeem Bilani
- Maroone Cancer Center, Cleveland Clinic Florida, Weston, Florida, USA
| | - Leah Elson
- Maroone Cancer Center, Cleveland Clinic Florida, Weston, Florida, USA
| | - Felipe Martinez
- Department of Radiology, Cleveland Clinic Florida, Weston, Florida, USA
| | - Diego Sadler
- Heart and Vascular Center, Cleveland Clinic Florida, Weston, Florida, USA
| | - Zeina Nahleh
- Maroone Cancer Center, Cleveland Clinic Florida, Weston, Florida, USA
| | | | - Evan Alley
- Maroone Cancer Center, Cleveland Clinic Florida, Weston, Florida, USA
| |
Collapse
|