1
|
Yang X, Hu J, Gao Q, Deng Y, Liu Y, He X, Li C, Yu X, Wan Y, Pi C, Wei Y, Li C. Advances in nano-delivery systems based on diagnosis and theranostics strategy for atherosclerosis. J Drug Target 2025; 33:492-507. [PMID: 39601425 DOI: 10.1080/1061186x.2024.2433560] [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: 08/05/2024] [Revised: 10/21/2024] [Accepted: 11/17/2024] [Indexed: 11/29/2024]
Abstract
Atherosclerosis (AS) is a chronic systemic inflammatory disease, where early diagnosis and theranostics strategy for AS are crucial for improving outcomes. However, conventional diagnostic techniques are limited in identifying early AS lesions, failing to stop the progression of AS in time. Nano-delivery systems have shown significant potential in AS diagnosis and treatment, offering distinct advantages in plaque identification and enhancing drugs concentration at lesion sites, thereby advancing new-generation theranostics strategy. This review discusses the application of nano-delivery systems based on imaging technology in AS diagnosis, and we further explore recent advancements in combining different imaging technologies with emerging theranostics strategy. In addition, we also discuss the challenges faced by nano-delivery systems for AS diagnosis and theranostics in clinical translation, such as nanoparticle targeting efficiency, cytotoxicity and long-term accumulation, immune clearance and inaccurate disease modelling. Finally, we also provide prospects on nano-delivery systems based on diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Xi Yang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Jian Hu
- Department of Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Quanle Gao
- Department of Neurology, Geriatrics, Hejiang County People's Hospital, Luzhou, Sichuan, China
| | - Yiping Deng
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yilin Liu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Xinghui He
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Chuang Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Xin Yu
- Chinese Pharmacy Laboratory, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Ying Wan
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Chao Pi
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yumeng Wei
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
2
|
Wu P, Xu L, Wang Q, Ma X, Wang X, Wang H, He S, Ru H, Zhao Y, Xiao Y, Zhang J, Wang X, An S, Hacker M, Li X, Zhang X, Wang Y, Yang M, Wu Z, Li S. Left Ventricular Remodelling Associated with the Transient Elevated [ 68Ga]Ga-Pentixafor Activity in the Remote Myocardium Following Acute Myocardial Infarction. Mol Imaging Biol 2024; 26:693-703. [PMID: 38641708 DOI: 10.1007/s11307-024-01912-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 02/17/2024] [Accepted: 03/14/2024] [Indexed: 04/21/2024]
Abstract
BACKGROUND Previous studies have initially reported accompanying elevated 2-deoxy-2[18F]fluoro-D-glucose ([18F]F-FDG) inflammatory activity in the remote area and its prognostic value after acute myocardial infarction (AMI). Non-invasive characterization of the accompanying inflammation in the remote myocardium may be of potency in guiding future targeted theranostics. [68Ga]Ga-Pentixafor targeting chemokine receptor 4 (CXCR4) on the surface of inflammatory cells is currently one of the promising inflammatory imaging agents. In this study, we sought to focus on the longitudinal evolution of [68Ga]Ga-Pentixafor activities in the remote myocardium following AMI and its association with cardiac function. METHODS Twelve AMI rats and six Sham rats serially underwent [68Ga]Ga-Pentixafor imaging at pre-operation, and 5, 7, 14 days post-operation. Maximum and mean standard uptake value (SUV) and target-to-background ratio (TBR) were assessed to indicate the uptake intensity. Gated [18F]F-FDG imaging and immunofluorescent staining were performed to obtain cardiac function and responses of pro-inflammatory and reparative macrophages, respectively. RESULTS The uptake of [68Ga]Ga-Pentixafor in the infarcted myocardium peaked at day 5 (all P = 0.003), retained at day 7 (all P = 0.011), and recovered at day 14 after AMI (P > 0.05), paralleling with the rise-fall pro-inflammatory M1 macrophages (P < 0.05). Correlated with the peak activity in the infarct territory, [68Ga]Ga-Pentixafor uptake in the remote myocardium on day 5 early after AMI significantly increased (AMI vs. Sham: SUVmean, SUVmax, and TBRmean: all P < 0.05), and strongly correlated with contemporaneous EDV and/or ESV (SUVmean and TBRmean: both P < 0.05). The transitory remote activity recovered as of day 7 post-AMI (AMI vs. Sham: P > 0.05). CONCLUSIONS Corresponding with the peaked [68Ga]Ga-Pentixafor activity in the infarcted myocardium, the activity in the remote region elevated accordingly and led to contemporaneous left ventricular remodelling early after AMI. Further studies are warranted to clarify its clinical application potential.
Collapse
Affiliation(s)
- Ping Wu
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, China
| | - Li Xu
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, China
| | - Qi Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Shanxi Medical University, Taiyuan, China
| | - Xiaofang Ma
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China
| | - Xinzhu Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Shanxi Medical University, Taiyuan, China
| | - Hongliang Wang
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, China
| | - Sheng He
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Huibin Ru
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, China
| | - Yuting Zhao
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China
| | - Yuxin Xiao
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China
| | - Jingying Zhang
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China
| | - Xinchao Wang
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, China
| | - Shaohui An
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, China
- Shanghai United Imaging Healthcare Co., Ltd., Shanghai, China
| | - Marcus Hacker
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Xiang Li
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Xiaoli Zhang
- Laboratory for Molecular Imaging, Department of Nuclear Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yuetao Wang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Minfu Yang
- Department of Nuclear Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Zhifang Wu
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, China
| | - Sijin Li
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China.
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
3
|
Tarchi SM, Salvatore M, Lichtenstein P, Sekar T, Capaccione K, Luk L, Shaish H, Makkar J, Desperito E, Leb J, Navot B, Goldstein J, Laifer S, Beylergil V, Ma H, Jambawalikar S, Aberle D, D'Souza B, Bentley-Hibbert S, Marin MP. Radiology of fibrosis part III: genitourinary system. J Transl Med 2024; 22:616. [PMID: 38961396 PMCID: PMC11223291 DOI: 10.1186/s12967-024-05333-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/20/2024] [Indexed: 07/05/2024] Open
Abstract
Fibrosis is a pathological process involving the abnormal deposition of connective tissue, resulting from improper tissue repair in response to sustained injury caused by hypoxia, infection, or physical damage. It can impact any organ, leading to their dysfunction and eventual failure. Additionally, tissue fibrosis plays an important role in carcinogenesis and the progression of cancer.Early and accurate diagnosis of organ fibrosis, coupled with regular surveillance, is essential for timely disease-modifying interventions, ultimately reducing mortality and enhancing quality of life. While extensive research has already been carried out on the topics of aberrant wound healing and fibrogenesis, we lack a thorough understanding of how their relationship reveals itself through modern imaging techniques.This paper focuses on fibrosis of the genito-urinary system, detailing relevant imaging technologies used for its detection and exploring future directions.
Collapse
Affiliation(s)
- Sofia Maria Tarchi
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA.
| | - Mary Salvatore
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Philip Lichtenstein
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Thillai Sekar
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Kathleen Capaccione
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Lyndon Luk
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Hiram Shaish
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Jasnit Makkar
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Elise Desperito
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Jay Leb
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Benjamin Navot
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Jonathan Goldstein
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Sherelle Laifer
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Volkan Beylergil
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Hong Ma
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Sachin Jambawalikar
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Dwight Aberle
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Belinda D'Souza
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Stuart Bentley-Hibbert
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Monica Pernia Marin
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| |
Collapse
|
4
|
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
|
5
|
Konrad M, Rinscheid A, Wienand G, Nittbaur B, Wester HJ, Janzen T, Lapa C, Pfob CH, Schottelius M. [ 99mTc]Tc-PentixaTec: development, extensive pre-clinical evaluation, and first human experience. Eur J Nucl Med Mol Imaging 2023; 50:3937-3948. [PMID: 37597009 PMCID: PMC10611619 DOI: 10.1007/s00259-023-06395-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 08/05/2023] [Indexed: 08/21/2023]
Abstract
PURPOSE The clinical success non-invasive imaging of CXCR4 expression using [68 Ga]Ga-PentixaFor-PET warrants an expansion of the targeting concept towards conventional scintigraphy/SPECT with their lower cost and general availability. To this aim, we developed and comparatively evaluated a series of 99mTc-labeled cyclic pentapeptides based on the PentixaFor scaffold. METHODS Six mas3-conjugated CPCR4 analogs with different 4-aminobenzoic acid (Abz)-D-Ala-D-Arg-aa3 linkers (L1-L6) as well as the corresponding HYNIC- and N4-analogs of L6-CPCR4 were synthesized via standard SPPS. Competitive binding studies (IC50 and IC50inv) were carried out using Jurkat T cell lymphoma cells and [125I]FC-131 as radioligand. Internalization kinetics were investigated using hCXCR4-overexpressing Chem-1 cells. Biodistribution studies and small animal SPECT/CT imaging (1 h p.i.) were carried out using Jurkat xenograft bearing CB17/SCID mice. Based on the preclinical results, [99mTc]Tc-N4-L6-CPCR4 ([99mTc]Tc-PentixaTec) was selected for an early translation to the human setting. Five patients with hematologic malignancies underwent [99mTc]Tc-N4-L6-CPCR4 SPECT/planar imaging with individual dosimetry. RESULTS Of the six mas3-conjugated peptides, mas3-L6-CPCR4 (mas3-dap-r-a-Abz-CPCR4) showed the highest CXCR4 affinity (IC50 = 5.0 ± 1.3 nM). Conjugation with N4 (N4-L6-CPCR4) further improved hCXCR4 affinity to 0.6 ± 0.1 nM. [99mTc]Tc-N4-L6-CPCR4 also showed the most efficient internalization (97% of total cellular activity at 2 h) and the highest tumor accumulation (8.6 ± 1.3% iD/g, 1 h p.i.) of the compounds investigated. Therefore, [99mTc]Tc-N4-L6-CPCR4 (termed [99mTc]Tc-PentixaTec) was selected for first-in-human application. [99mTc]Tc-PentixaTec was well tolerated, exhibits a favorable biodistribution and dosimetry profile (2.1-3.4 mSv per 500 MBq) and excellent tumor/background ratios in SPECT and planar imaging. CONCLUSION The successive optimization of the amino acid composition of the linker structure and the N-terminal 99mTc-labeling strategies (mas3 vs HYNIC vs N4) has provided [99mTc]Tc-PentixaTec as a novel, highly promising CXCR4-targeted SPECT agent for clinical application. With its excellent CXCR4 affinity, efficient internalization, high uptake in CXCR4-expressing tissues, suitable clearance/biodistribution characteristics, and favorable human dosimetry, it holds great potential for further clinical use.
Collapse
Affiliation(s)
- Matthias Konrad
- Chair for Pharmaceutical Radiochemistry, Faculties of Chemistry and Medicine, Technische Universität München, 85748, Garching, Germany
| | - Andreas Rinscheid
- Medical Physics and Radiation Protection, University Hospital Augsburg, Stenglinstrasse 2, 86156, Augsburg, Germany
| | - Georgine Wienand
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Stenglinstrasse 2, 86156, Augsburg, Germany
| | - Bernd Nittbaur
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Stenglinstrasse 2, 86156, Augsburg, Germany
| | - Hans-Jürgen Wester
- Chair for Pharmaceutical Radiochemistry, Faculties of Chemistry and Medicine, Technische Universität München, 85748, Garching, Germany
| | - Tilman Janzen
- Medical Physics and Radiation Protection, University Hospital Augsburg, Stenglinstrasse 2, 86156, Augsburg, Germany
| | - Constantin Lapa
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Stenglinstrasse 2, 86156, Augsburg, Germany
| | - Christian Helmut Pfob
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Stenglinstrasse 2, 86156, Augsburg, Germany.
| | - Margret Schottelius
- Translational Radiopharmaceutical Sciences, Department of Nuclear Medicine and Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Rue du Bugnon 25A, Agora, CH-1011, Lausanne, Switzerland.
- AGORA, Pôle de Recherche Sur Le Cancer, 1011, Lausanne, Switzerland.
- SCCL Swiss Cancer Center Leman, 1011, Lausanne, Switzerland.
| |
Collapse
|
6
|
Mézquita AJV, Biavati F, Falk V, Alkadhi H, Hajhosseiny R, Maurovich-Horvat P, Manka R, Kozerke S, Stuber M, Derlin T, Channon KM, Išgum I, Coenen A, Foellmer B, Dey D, Volleberg RHJA, Meinel FG, Dweck MR, Piek JJ, van de Hoef T, Landmesser U, Guagliumi G, Giannopoulos AA, Botnar RM, Khamis R, Williams MC, Newby DE, Dewey M. Clinical quantitative coronary artery stenosis and coronary atherosclerosis imaging: a Consensus Statement from the Quantitative Cardiovascular Imaging Study Group. Nat Rev Cardiol 2023; 20:696-714. [PMID: 37277608 DOI: 10.1038/s41569-023-00880-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/19/2023] [Indexed: 06/07/2023]
Abstract
The detection and characterization of coronary artery stenosis and atherosclerosis using imaging tools are key for clinical decision-making in patients with known or suspected coronary artery disease. In this regard, imaging-based quantification can be improved by choosing the most appropriate imaging modality for diagnosis, treatment and procedural planning. In this Consensus Statement, we provide clinical consensus recommendations on the optimal use of different imaging techniques in various patient populations and describe the advances in imaging technology. Clinical consensus recommendations on the appropriateness of each imaging technique for direct coronary artery visualization were derived through a three-step, real-time Delphi process that took place before, during and after the Second International Quantitative Cardiovascular Imaging Meeting in September 2022. According to the Delphi survey answers, CT is the method of choice to rule out obstructive stenosis in patients with an intermediate pre-test probability of coronary artery disease and enables quantitative assessment of coronary plaque with respect to dimensions, composition, location and related risk of future cardiovascular events, whereas MRI facilitates the visualization of coronary plaque and can be used in experienced centres as a radiation-free, second-line option for non-invasive coronary angiography. PET has the greatest potential for quantifying inflammation in coronary plaque but SPECT currently has a limited role in clinical coronary artery stenosis and atherosclerosis imaging. Invasive coronary angiography is the reference standard for stenosis assessment but cannot characterize coronary plaques. Finally, intravascular ultrasonography and optical coherence tomography are the most important invasive imaging modalities for the identification of plaques at high risk of rupture. The recommendations made in this Consensus Statement will help clinicians to choose the most appropriate imaging modality on the basis of the specific clinical scenario, individual patient characteristics and the availability of each imaging modality.
Collapse
Affiliation(s)
| | - Federico Biavati
- Department of Radiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Volkmar Falk
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research) Partner Site, Berlin, Germany
- Department of Health Science and Technology, ETH Zurich, Zurich, Switzerland
| | - Hatem Alkadhi
- Diagnostic and Interventional Radiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Reza Hajhosseiny
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Pál Maurovich-Horvat
- Department of Radiology, Medical Imaging Center, Semmelweis University, Budapest, Hungary
| | - Robert Manka
- Diagnostic and Interventional Radiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland
| | - Sebastian Kozerke
- Institute for Biomedical Engineering, ETH Zurich, University of Zurich, Zurich, Switzerland
| | - Matthias Stuber
- Department of Radiology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Keith M Channon
- Radcliffe Department of Medicine, University of Oxford and Oxford University Hospitals, Oxford, UK
| | - Ivana Išgum
- Department of Biomedical Engineering and Physics, Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Adriaan Coenen
- Department of Radiology, Erasmus University, Rotterdam, Netherlands
| | - Bernhard Foellmer
- Department of Radiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Damini Dey
- Departments of Biomedical Sciences and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Rick H J A Volleberg
- Department of Cardiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Felix G Meinel
- Department of Radiology, University Medical Centre Rostock, Rostock, Germany
| | - Marc R Dweck
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Jan J Piek
- Department of Clinical and Experimental Cardiology and Cardiovascular Sciences, Amsterdam UMC, Heart Center, University of Amsterdam, Amsterdam, Netherlands
| | - Tim van de Hoef
- Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Ulf Landmesser
- DZHK (German Centre for Cardiovascular Research) Partner Site, Berlin, Germany
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Giulio Guagliumi
- Division of Cardiology, IRCCS Galeazzi Sant'Ambrogio Hospital, Milan, Italy
| | - Andreas A Giannopoulos
- Department of Nuclear Medicine, Cardiac Imaging, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - René M Botnar
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
- Institute for Biological and Medical Engineering, Pontificia Universidad Católica de Chile, Millennium Institute for Intelligent Healthcare Engineering, Santiago, Chile
| | - Ramzi Khamis
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - David E Newby
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Marc Dewey
- Department of Radiology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research) Partner Site, Berlin, Germany.
- Deutsches Herzzentrum der Charité (DHZC), Charité - Universitätsmedizin Berlin, Berlin, Germany.
- Berlin Institute of Health, Campus Charité Mitte, Berlin, Germany.
| |
Collapse
|
7
|
Soni SS, D'Elia AM, Rodell CB. Control of the post-infarct immune microenvironment through biotherapeutic and biomaterial-based approaches. Drug Deliv Transl Res 2023; 13:1983-2014. [PMID: 36763330 PMCID: PMC9913034 DOI: 10.1007/s13346-023-01290-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2023] [Indexed: 02/11/2023]
Abstract
Ischemic heart failure (IHF) is a leading cause of morbidity and mortality worldwide, for which heart transplantation remains the only definitive treatment. IHF manifests from myocardial infarction (MI) that initiates tissue remodeling processes, mediated by mechanical changes in the tissue (loss of contractility, softening of the myocardium) that are interdependent with cellular mechanisms (cardiomyocyte death, inflammatory response). The early remodeling phase is characterized by robust inflammation that is necessary for tissue debridement and the initiation of repair processes. While later transition toward an immunoregenerative function is desirable, functional reorientation from an inflammatory to reparatory environment is often lacking, trapping the heart in a chronically inflamed state that perpetuates cardiomyocyte death, ventricular dilatation, excess fibrosis, and progressive IHF. Therapies can redirect the immune microenvironment, including biotherapeutic and biomaterial-based approaches. In this review, we outline these existing approaches, with a particular focus on the immunomodulatory effects of therapeutics (small molecule drugs, biomolecules, and cell or cell-derived products). Cardioprotective strategies, often focusing on immunosuppression, have shown promise in pre-clinical and clinical trials. However, immunoregenerative therapies are emerging that often benefit from exacerbating early inflammation. Biomaterials can be used to enhance these therapies as a result of their intrinsic immunomodulatory properties, parallel mechanisms of action (e.g., mechanical restraint), or by enabling cell or tissue-targeted delivery. We further discuss translatability and the continued progress of technologies and procedures that contribute to the bench-to-bedside development of these critically needed treatments.
Collapse
Affiliation(s)
- Shreya S Soni
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Arielle M D'Elia
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Christopher B Rodell
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA.
| |
Collapse
|
8
|
Nappi C, Panico M, Falzarano M, Vallone C, Ponsiglione A, Cutillo P, Zampella E, Petretta M, Cuocolo A. Tracers for Cardiac Imaging: Targeting the Future of Viable Myocardium. Pharmaceutics 2023; 15:pharmaceutics15051532. [PMID: 37242772 DOI: 10.3390/pharmaceutics15051532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/02/2023] [Accepted: 05/07/2023] [Indexed: 05/28/2023] Open
Abstract
Ischemic heart disease is the leading cause of mortality worldwide. In this context, myocardial viability is defined as the amount of myocardium that, despite contractile dysfunction, maintains metabolic and electrical function, having the potential for functional enhancement upon revascularization. Recent advances have improved methods to detect myocardial viability. The current paper summarizes the pathophysiological basis of the current methods used to detect myocardial viability in light of the advancements in the development of new radiotracers for cardiac imaging.
Collapse
Affiliation(s)
- Carmela Nappi
- Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131 Naples, Italy
| | - Mariarosaria Panico
- Institute of Biostructure and Bioimaging, National Council of Research, 80131 Naples, Italy
| | - Maria Falzarano
- Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131 Naples, Italy
| | - Carlo Vallone
- Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131 Naples, Italy
| | - Andrea Ponsiglione
- Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131 Naples, Italy
| | - Paolo Cutillo
- Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131 Naples, Italy
| | - Emilia Zampella
- Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131 Naples, Italy
| | - Mario Petretta
- IRCCS SYNLAB SDN, Via Gianturco 113, 80131 Naples, Italy
| | - Alberto Cuocolo
- Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
9
|
Abohashem S, Grewal SS, Tawakol A, Osborne MT. Radionuclide Imaging of Heart-Brain Connections. Cardiol Clin 2023; 41:267-275. [PMID: 37003682 PMCID: PMC10152492 DOI: 10.1016/j.ccl.2023.01.013] [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] [Indexed: 02/21/2023]
Abstract
The heart and brain have a complex interplay wherein disease or injury to either organ may adversely affect the other. The mechanisms underlying this connection remain incompletely characterized. However, nuclear molecular imaging is uniquely suited to investigate these pathways by facilitating the simultaneous assessment of both organs using targeted radiotracers. Research within this paradigm has demonstrated important roles for inflammation, autonomic nervous system and neurohormonal activity, metabolism, and perfusion in the heart-brain connection. Further mechanistic clarification may facilitate greater clinical awareness and the development of targeted therapies to alleviate the burden of disease in both organs.
Collapse
Affiliation(s)
- Shady Abohashem
- Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA; Massachusetts General Hospital, Cardiovascular Imaging Research Center, 165 Cambridge Street, Suite 400, Boston, MA 02114, USA
| | - Simran S Grewal
- Massachusetts General Hospital, Cardiovascular Imaging Research Center, 165 Cambridge Street, Suite 400, Boston, MA 02114, USA; Division of Cardiology, Department of Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
| | - Ahmed Tawakol
- Massachusetts General Hospital, Cardiovascular Imaging Research Center, 165 Cambridge Street, Suite 400, Boston, MA 02114, USA; Division of Cardiology, Department of Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
| | - Michael T Osborne
- Massachusetts General Hospital, Cardiovascular Imaging Research Center, 165 Cambridge Street, Suite 400, Boston, MA 02114, USA; Division of Cardiology, Department of Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA.
| |
Collapse
|
10
|
[ 68Ga]Ga-NODAGA-E[(cRGDyK)] 2 and [ 64Cu]Cu-DOTATATE PET Predict Improvement in Ischemic Cardiomyopathy. Diagnostics (Basel) 2023; 13:diagnostics13020268. [PMID: 36673078 PMCID: PMC9857952 DOI: 10.3390/diagnostics13020268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/05/2023] [Accepted: 01/07/2023] [Indexed: 01/12/2023] Open
Abstract
An increasing number of patients are living with chronic ischemic cardiomyopathy (ICM) and/or heart failure. Treatment options and prognostic tools are lacking for many of these patients. Our aim was to investigate the prognostic value of imaging angiogenesis and macrophage activation via positron emission tomography (PET) in terms of functional improvement after cell therapy. Myocardial infarction was induced in rats. Animals were scanned with [18F]FDG PET and echocardiography after four weeks and randomized to allogeneic adipose tissue-derived stromal cells (ASCs, n = 18) or saline (n = 9). Angiogenesis and macrophage activation were assessed before and after treatment by [68Ga]Ga-RGD and [64Cu]Cu-DOTATATE. There was no overall effect of the treatment. Rats that improved left ventricular ejection fraction (LVEF) had higher uptake of both [68Ga]Ga-RGD and [64Cu]Cu-DOTATATE at follow-up (p = 0.006 and p = 0.008, respectively). The uptake of the two tracers correlated with each other (r = 0.683, p = 0.003 pre-treatment and r = 0.666, p = 0.004 post-treatment). SUVmax at follow-up could predict improvement in LVEF (p = 0.016 for [68Ga]Ga-RGD and p = 0.045 for [64Cu]Cu-DOTATATE). High uptake of [68Ga]Ga-RGD and [64Cu]Cu-DOTATATE PET after injection of ASCs or saline preceded improvement in LVEF. The use of these tracers could improve the monitoring of heart failure patients in treatment.
Collapse
|
11
|
Recent Advances in Cardiovascular Diseases Research Using Animal Models and PET Radioisotope Tracers. Int J Mol Sci 2022; 24:ijms24010353. [PMID: 36613797 PMCID: PMC9820417 DOI: 10.3390/ijms24010353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
Cardiovascular diseases (CVD) is a collective term describing a range of conditions that affect the heart and blood vessels. Due to the varied nature of the disorders, distinguishing between their causes and monitoring their progress is crucial for finding an effective treatment. Molecular imaging enables non-invasive visualisation and quantification of biological pathways, even at the molecular and subcellular levels, what is essential for understanding the causes and development of CVD. Positron emission tomography imaging is so far recognized as the best method for in vivo studies of the CVD related phenomena. The imaging is based on the use of radioisotope-labelled markers, which have been successfully used in both pre-clinical research and clinical studies. Current research on CVD with the use of such radioconjugates constantly increases our knowledge and understanding of the causes, and brings us closer to effective monitoring and treatment. This review outlines recent advances in the use of the so-far available radioisotope markers in the research on cardiovascular diseases in rodent models, points out the problems and provides a perspective for future applications of PET imaging in CVD studies.
Collapse
|
12
|
Park J, Young BD, Miller EJ. Potential novel imaging targets of inflammation in cardiac sarcoidosis. J Nucl Cardiol 2022; 29:2171-2187. [PMID: 34734365 DOI: 10.1007/s12350-021-02838-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 09/26/2021] [Indexed: 10/19/2022]
Abstract
Cardiac sarcoidosis (CS) is an inflammatory disease with high morbidity and mortality, with a pathognomonic feature of non-caseating granulomatous inflammation. While 18F-fluorodeoxyglucose (FDG) positron emission tomography (PET) is a well-established modality to image inflammation and diagnose CS, there are limitations to its specificity and reproducibility. Imaging focused on the molecular processes of inflammation including the receptors and cellular microenvironments present in sarcoid granulomas provides opportunities to improve upon FDG-PET imaging for CS. This review will highlight the current limitations of FDG-PET imaging for CS while discussing emerging new nuclear imaging molecular targets for the imaging of cardiac sarcoidosis.
Collapse
Affiliation(s)
- Jakob Park
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Bryan D Young
- Section of Cardiovascular Medicine, Department of Medicine, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Edward J Miller
- Section of Cardiovascular Medicine, Department of Medicine, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA.
| |
Collapse
|
13
|
Teng YY, Zou ML, Liu SY, Jia Y, Zhang KW, Yuan ZD, Wu JJ, Ye JX, Yu S, Li X, Zhou XJ, Yuan FL. Dual-Action Icariin-Containing Thermosensitive Hydrogel for Wound Macrophage Polarization and Hair-Follicle Neogenesis. Front Bioeng Biotechnol 2022; 10:902894. [PMID: 35832407 PMCID: PMC9272914 DOI: 10.3389/fbioe.2022.902894] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 06/03/2022] [Indexed: 01/01/2023] Open
Abstract
Bone morphogenetic protein (BMP) pathway is essential for M2 macrophage polarization and hair-follicle neogenesis. Icariin, a flavonoid derived from Epimedium, is a mediator of the BMP pathway. Here, we develop a hydrogel formulation functionalized with icariin for regulation of macrophage polarization to accelerate wound healing and hair-follicle neogenesis. Compared to skin defects without icariin treatment, those treated with icariin+PEG hydrogel healed faster and had new hair follicles. Results in vivo showed that icariin+PEG hydrogel induced a higher level of M2 phenotypic transformation of macrophages. Moreover, icariin+PEG hydrogel significantly accelerated wound-repair process by reducing the invasion of inflammation, excessive deposition of collagen, immoderate activation of myofibroblasts, and increasing the regeneration of hair follicles. Furthermore, studies in vitro demonstrated that the icariin+PEG hydrogel induced macrophages to polarize to the M2 phenotype and dermal papilla cell to hair follicles. Finally, molecular analysis demonstrated that the icariin+PEG hydrogel increased the expression of BMP4 and Smad1/5 phosphorylation in skin wounds. These results demonstrate the therapeutic potential of icariin-containing thermosensitive hydrogels for inducing M2 macrophage polarization to accelerate wound healing and promote hair-follicle neogenesis by regulating the BMP pathway.
Collapse
Affiliation(s)
- Ying-Ying Teng
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, China
| | - Ming-Li Zou
- Wuxi Clinical Medicine School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Wuxi, China
| | - Si-Yu Liu
- Wuxi Clinical Medicine School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Wuxi, China
| | - Yuan Jia
- Wuxi Clinical Medicine School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Wuxi, China
| | - Kai-Wen Zhang
- Wuxi Clinical Medicine School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Wuxi, China
| | - Zheng-Dong Yuan
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, China
| | - Jun-Jie Wu
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, China
| | - Jun-Xing Ye
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, China
| | - Shun Yu
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, China
| | - Xia Li
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, China
| | - Xiao-Jin Zhou
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, China
| | - Feng-Lai Yuan
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, China.,Wuxi Clinical Medicine School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Wuxi, China
| |
Collapse
|
14
|
Wilk B, Smailovic H, Wisenberg G, Sykes J, Butler J, Kovacs M, Thiessen JD, Prato FS. Tracking the progress of inflammation with PET/MRI in a canine model of myocardial infarction. J Nucl Cardiol 2022; 29:1315-1325. [PMID: 33462785 DOI: 10.1007/s12350-020-02487-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 11/28/2020] [Indexed: 11/25/2022]
Abstract
BACKGROUND Following myocardial infarction, tissue undergoes pathophysiological changes involving inflammation and scar tissue formation. However, little is known about the pathophysiology and prognostic significance of any corresponding changes in remote myocardium. The aim of this study was to investigate the potential application of a combined constant infusion of 18F-FDG and Gd-DTPA to quantitate inflammation and extracellular volume (ECV) from 3 to 40 days after myocardial infarction. METHODS Eight canine subjects were imaged at multiple time points following induction of an MI with a 60-minute concurrent constant infusion of Gd-DTPA and 18F-FDG using a hybrid PET/MRI scanner. RESULTS There was a significant increase in ECV in remote myocardium on day 14 post-MI (P = .034) and day 21 (P = .021) compared to the baseline. ECV was significantly elevated in the infarcted myocardium compared to remote myocardium at all time points post-MI (days 3, 7, 14, 21, and 40) (P < .001) while glucose uptake was also increased within the infarct on days 3, 7, 14, and 21 but not 40. CONCLUSIONS The significant increase in ECV in remote tissue may be due to an ongoing inflammatory process in the early weeks post-infarct.
Collapse
Affiliation(s)
- B Wilk
- Lawson Health Research Institute, London, Canada.
- Department of Medical Biophysics, Western University, London, Canada.
- Collaborative Graduate Program in Molecular Imaging, Western University, London, Canada.
| | - H Smailovic
- Lawson Health Research Institute, London, Canada
- Department of Medical Imaging, Western University, London, Canada
| | - G Wisenberg
- Lawson Health Research Institute, London, Canada
- MyHealth Centre, Arva, Canada
| | - J Sykes
- Lawson Health Research Institute, London, Canada
| | - J Butler
- Lawson Health Research Institute, London, Canada
| | - M Kovacs
- Lawson Health Research Institute, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
- Department of Medical Imaging, Western University, London, Canada
| | - J D Thiessen
- Lawson Health Research Institute, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
- Department of Medical Imaging, Western University, London, Canada
- Collaborative Graduate Program in Molecular Imaging, Western University, London, Canada
| | - F S Prato
- Lawson Health Research Institute, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
- Department of Medical Imaging, Western University, London, Canada
- Collaborative Graduate Program in Molecular Imaging, Western University, London, Canada
| |
Collapse
|
15
|
Toner YC, Ghotbi AA, Naidu S, Sakurai K, van Leent MMT, Jordan S, Ordikhani F, Amadori L, Sofias AM, Fisher EL, Maier A, Sullivan N, Munitz J, Senders ML, Mason C, Reiner T, Soultanidis G, Tarkin JM, Rudd JHF, Giannarelli C, Ochando J, Pérez-Medina C, Kjaer A, Mulder WJM, Fayad ZA, Calcagno C. Systematically evaluating DOTATATE and FDG as PET immuno-imaging tracers of cardiovascular inflammation. Sci Rep 2022; 12:6185. [PMID: 35418569 PMCID: PMC9007951 DOI: 10.1038/s41598-022-09590-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 03/22/2022] [Indexed: 02/08/2023] Open
Abstract
In recent years, cardiovascular immuno-imaging by positron emission tomography (PET) has undergone tremendous progress in preclinical settings. Clinically, two approved PET tracers hold great potential for inflammation imaging in cardiovascular patients, namely FDG and DOTATATE. While the former is a widely applied metabolic tracer, DOTATATE is a relatively new PET tracer targeting the somatostatin receptor 2 (SST2). In the current study, we performed a detailed, head-to-head comparison of DOTATATE-based radiotracers and [18F]F-FDG in mouse and rabbit models of cardiovascular inflammation. For mouse experiments, we labeled DOTATATE with the long-lived isotope [64Cu]Cu to enable studying the tracer's mode of action by complementing in vivo PET/CT experiments with thorough ex vivo immunological analyses. For translational PET/MRI rabbit studies, we employed the more widely clinically used [68Ga]Ga-labeled DOTATATE, which was approved by the FDA in 2016. DOTATATE's pharmacokinetics and timed biodistribution were determined in control and atherosclerotic mice and rabbits by ex vivo gamma counting of blood and organs. Additionally, we performed in vivo PET/CT experiments in mice with atherosclerosis, mice subjected to myocardial infarction and control animals, using both [64Cu]Cu-DOTATATE and [18F]F-FDG. To evaluate differences in the tracers' cellular specificity, we performed ensuing ex vivo flow cytometry and gamma counting. In mice subjected to myocardial infarction, in vivo [64Cu]Cu-DOTATATE PET showed higher differential uptake between infarcted (SUVmax 1.3, IQR, 1.2-1.4, N = 4) and remote myocardium (SUVmax 0.7, IQR, 0.5-0.8, N = 4, p = 0.0286), and with respect to controls (SUVmax 0.6, IQR, 0.5-0.7, N = 4, p = 0.0286), than [18F]F-FDG PET. In atherosclerotic mice, [64Cu]Cu-DOTATATE PET aortic signal, but not [18F]F-FDG PET, was higher compared to controls (SUVmax 1.1, IQR, 0.9-1.3 and 0.5, IQR, 0.5-0.6, respectively, N = 4, p = 0.0286). In both models, [64Cu]Cu-DOTATATE demonstrated preferential accumulation in macrophages with respect to other myeloid cells, while [18F]F-FDG was taken up by macrophages and other leukocytes. In a translational PET/MRI study in atherosclerotic rabbits, we then compared [68Ga]Ga-DOTATATE and [18F]F-FDG for the assessment of aortic inflammation, combined with ex vivo radiometric assays and near-infrared imaging of macrophage burden. Rabbit experiments showed significantly higher aortic accumulation of both [68Ga]Ga-DOTATATE and [18F]F-FDG in atherosclerotic (SUVmax 0.415, IQR, 0.338-0.499, N = 32 and 0.446, IQR, 0.387-0.536, N = 27, respectively) compared to control animals (SUVmax 0.253, IQR, 0.197-0.285, p = 0.0002, N = 10 and 0.349, IQR, 0.299-0.423, p = 0.0159, N = 11, respectively). In conclusion, we present a detailed, head-to-head comparison of the novel SST2-specific tracer DOTATATE and the validated metabolic tracer [18F]F-FDG for the evaluation of inflammation in small animal models of cardiovascular disease. Our results support further investigations on the use of DOTATATE to assess cardiovascular inflammation as a complementary readout to the widely used [18F]F-FDG.
Collapse
Affiliation(s)
- Yohana C Toner
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Ave, PO Box: 1234, New York, NY, 10029, USA
- Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Adam A Ghotbi
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Ave, PO Box: 1234, New York, NY, 10029, USA
- Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Clinical Physiology, Nuclear Medicine and PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Sonum Naidu
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Ave, PO Box: 1234, New York, NY, 10029, USA
- Diagnostic, Molecular and Interventional Radiology, 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, 1470 Madison Ave, PO Box: 1234, New York, NY, 10029, USA
- 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, 1470 Madison Ave, PO Box: 1234, New York, NY, 10029, USA
- Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stefan Jordan
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Institute of Microbiology, Infectious Diseases and Immunology, Berlin, Germany
| | - Farideh Ordikhani
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Letizia Amadori
- Department of Genetics and Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- New York University Cardiovascular Research Center, Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine, New York University Langone Health, New York, NY, USA
| | - Alexandros Marios Sofias
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Ave, PO Box: 1234, New York, NY, 10029, USA
- Diagnostic, Molecular and Interventional Radiology, 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 (NTNU), Trondheim, Norway
| | - Elizabeth L Fisher
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Ave, PO Box: 1234, New York, NY, 10029, USA
- Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexander Maier
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Ave, PO Box: 1234, New York, NY, 10029, USA
- Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Cardiology and Angiology I, Faculty of Medicine, Heart Center Freiburg University, University of Freiburg, Freiburg, Germany
| | - Nathaniel Sullivan
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Ave, PO Box: 1234, New York, NY, 10029, USA
- Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jazz Munitz
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Ave, PO Box: 1234, New York, NY, 10029, USA
- Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Max L Senders
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Ave, PO Box: 1234, New York, NY, 10029, USA
- Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Christian Mason
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Department of Radiology and Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA
| | - Georgios Soultanidis
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Ave, PO Box: 1234, New York, NY, 10029, USA
- Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jason M Tarkin
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - James H F Rudd
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Chiara Giannarelli
- Department of Genetics and Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- New York University Cardiovascular Research Center, Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine, New York University Langone Health, New York, NY, USA
- Cardiovascular Research Center, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jordi Ochando
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Transplant Immunology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos Pérez-Medina
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Ave, PO Box: 1234, New York, NY, 10029, USA
- Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine and PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Willem J M Mulder
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Ave, PO Box: 1234, New York, NY, 10029, USA
- Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- Laboratory of Chemical Biology, Department of Biochemical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Zahi A Fayad
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Ave, PO Box: 1234, New York, NY, 10029, USA
- Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Claudia Calcagno
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Ave, PO Box: 1234, New York, NY, 10029, USA.
- Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
16
|
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
|
17
|
Teiler J, Ahl M, Åkerlund B, Brismar H, Holstensson M, Gabrielson S, Hedlund H, Axelsson R. 99mTc-HMPAO-WBC SPECT/CT versus 18F-FDG-WBC PET/CT in chronic prosthetic joint infection: a pilot study. Nucl Med Commun 2022; 43:193-200. [PMID: 34678830 DOI: 10.1097/mnm.0000000000001502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE The aim of this study was to compare 99mTc-HMPAO-WBC-SPECT/CT combined with 99mTc-nanocollloid SPECT/CT and 18F-FDG-WBC-PET/CT combined with 99mTc-Nanocollloid SPECT/CT for the diagnosis and treatment evaluation of chronic prosthetic joint infection (PJI). METHODS Patients with suspected chronic PJI were examined with 99mTc-HMPAO-WBC SPECT/CT, 18F-FDG-WBC PET/CT, and 99mTc-nanocolloid SPECT/CT (to visualize bone marrow). The location and patterns of uptake were noted and compared between the two leukocyte examinations. Both leukocyte examinations were evaluated visually for infection. The PET examinations were also evaluated semiquantitatively. Chronic PJI was verified clinically by microbial culture and successfully treated PJI was confirmed by 12 months symptom-free follow-up after cessation of antibiotics. RESULTS Nineteen patients were included with 10 hip prostheses and nine knee prostheses. Fourteen were diagnosed with chronic PJI and five with successfully treated PJI. The sensitivity of visual evaluation of 99mTc-WBC-HMPAO SPECT/CT for all joints was 0.31 and for 18F-FDG-WBC PET/CT 0.38. The specificity was 0.80 and 0.83, respectively. All patients with a true-positive SPECT examination had a false-negative PET examination and vice versa. Semiquantitative evaluation of the hips gave an area under the curve of 0.905 using the iliac crest as the background. Semiquantitative evaluation of the knees did not produce significant results. CONCLUSION This pilot study showed no difference in the sensitivity or specificity of 99mTc-HMPAO-WBC SPECT/CT and 18F-FDG-WBC PET/CT when combined with 99mTc-nanocollloid SPECT/CT in the diagnosis or treatment evaluation of suspected late chronic PJI.
Collapse
Affiliation(s)
- Johan Teiler
- Division of Radiology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm
- Department of Radiology, Karolinska University Hospital, Huddinge
| | - Marcus Ahl
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm
- Unit of Infectious Diseases, Karolinska University Hospital, Huddinge
| | - Börje Åkerlund
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm
- Unit of Infectious Diseases, Karolinska University Hospital, Huddinge
| | - Harald Brismar
- Division of Orthopaedics and Biotechnology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm
- Department of Orthopaedic Surgery, Karolinska University Hospital, Huddinge
| | - Maria Holstensson
- Functional Imaging and Technology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm
- Function Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Huddinge, Sweden
| | - Stefan Gabrielson
- Division of Radiology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm
- Radiology Service, Christchurch Hospital, Christchurch, New Zealand
| | - Håkan Hedlund
- Division of Orthopaedics and Biotechnology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm
- Department of Orthopaedic Surgery, Visby Hospital, Visby, Sweden
| | - Rimma Axelsson
- Division of Radiology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm
- Functional Imaging and Technology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm
| |
Collapse
|
18
|
Molecular imaging of the brain-heart axis provides insights into cardiac dysfunction after cerebral ischemia. Basic Res Cardiol 2022; 117:52. [PMID: 36279013 PMCID: PMC9592646 DOI: 10.1007/s00395-022-00961-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/26/2022] [Accepted: 10/13/2022] [Indexed: 01/31/2023]
Abstract
Ischemic stroke imparts elevated risk of heart failure though the underlying mechanisms remain poorly described. We aimed to characterize the influence of cerebral ischemic injury on cardiac function using multimodality molecular imaging to investigate brain and cardiac morphology and tissue inflammation in two mouse models of variable stroke severity. Transient middle cerebral artery occlusion (MCAo) generated extensive stroke damage (56.31 ± 40.39 mm3). Positron emission tomography imaging of inflammation targeting the mitochondrial translocator protein (TSPO) revealed localized neuroinflammation at 7 days after stroke compared to sham (3.8 ± 0.8 vs 2.6 ± 0.7 %ID/g max, p < 0.001). By contrast, parenchyma topical application of vasoconstrictor endothelin-1 did not generate significant stroke damage or neuroinflammatory cell activity. MCAo evoked a modest reduction in left ventricle ejection fraction at both 1 weeks and 3 weeks after stroke (LVEF at 3 weeks: 54.3 ± 5.7 vs 66.1 ± 3.5%, p < 0.001). This contractile impairment was paralleled by elevated cardiac TSPO PET signal compared to sham (8.6 ± 2.4 vs 5.8 ± 0.7%ID/g, p = 0.022), but was independent of leukocyte infiltration defined by flow cytometry. Stroke size correlated with severity of cardiac dysfunction (r = 0.590, p = 0.008). Statistical parametric mapping identified a direct association between neuroinflammation at 7 days in a cluster of voxels including the insular cortex and reduced ejection fraction (ρ = - 0.396, p = 0.027). Suppression of microglia led to lower TSPO signal at 7 days which correlated with spared late cardiac function after MCAo (r = - 0.759, p = 0.029). Regional neuroinflammation early after cerebral ischemia influences subsequent cardiac dysfunction. Total body TSPO PET enables monitoring of neuroinflammation, providing insights into brain-heart inter-organ communication and may guide therapeutic intervention to spare cardiac function post-stroke.
Collapse
|
19
|
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: 13] [Impact Index Per Article: 4.3] [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
|
20
|
More S, Marakalala MJ, Sathekge M. Tuberculosis: Role of Nuclear Medicine and Molecular Imaging With Potential Impact of Neutrophil-Specific Tracers. Front Med (Lausanne) 2021; 8:758636. [PMID: 34957144 PMCID: PMC8703031 DOI: 10.3389/fmed.2021.758636] [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: 08/14/2021] [Accepted: 11/03/2021] [Indexed: 01/02/2023] Open
Abstract
With Tuberculosis (TB) affecting millions of people worldwide, novel imaging modalities and tools, particularly nuclear medicine and molecular imaging, have grown with greater interest to assess the biology of the tuberculous granuloma and evolution thereof. Much early work has been performed at the pre-clinical level using gamma single photon emission computed tomography (SPECT) agents exploiting certain characteristics of Mycobacterium tuberculosis (MTb). Both antituberculous SPECT and positron emission tomography (PET) agents have been utilised to characterise MTb. Other PET tracers have been utilised to help to characterise the biology of MTb (including Gallium-68-labelled radiopharmaceuticals). Of all the tracers, 2-[18F]FDG has been studied extensively over the last two decades in many aspects of the treatment paradigm of TB: at diagnosis, staging, response assessment, restaging, and in potentially predicting the outcome of patients with latent TB infection. Its lower specificity in being able to distinguish different inflammatory cell types in the granuloma has garnered interest in reviewing more specific agents that can portend prognostic implications in the management of MTb. With the neutrophil being a cell type that portends this poorer prognosis, imaging this cell type may be able to answer more accurately questions relating to the tuberculous granuloma transmissivity and may help in characterising patients who may be at risk of developing active TB. The formyl peptide receptor 1(FPR1) expressed by neutrophils is a key marker in this process and is a potential target to characterise these areas. The pre-clinical work regarding the role of radiolabelled N-cinnamoyl –F-(D) L – F – (D) –L F (cFLFLF) (which is an antagonist for FPR1) using Technetium 99m-labelled conjugates and more recently radiolabelled with Gallium-68 and Copper 64 is discussed. It is the hope that further work with this tracer may accelerate its potential to be utilised in responding to many of the current diagnostic dilemmas and challenges in TB management, thereby making the tracer a translatable option in routine clinical care.
Collapse
Affiliation(s)
- Stuart More
- Division of Nuclear Medicine, Department of Radiation Medicine, University of Cape Town, Cape Town, South Africa
- Department of Nuclear Medicine, University of Pretoria and Steve Biko Academic Hospital, Pretoria, South Africa
- Nuclear Medicine Research Infrastructure, Steve Biko Academic Hospital, Pretoria, South Africa
- *Correspondence: Stuart More
| | - Mohlopheni J. Marakalala
- Africa Health Research Institute, Durban, South Africa
- Division of Infection and Immunity, University College London, London, United Kingdom
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
- Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Michael Sathekge
- Department of Nuclear Medicine, University of Pretoria and Steve Biko Academic Hospital, Pretoria, South Africa
- Nuclear Medicine Research Infrastructure, Steve Biko Academic Hospital, Pretoria, South Africa
| |
Collapse
|
21
|
Schottelius M, Herrmann K, Lapa C. In Vivo Targeting of CXCR4-New Horizons. Cancers (Basel) 2021; 13:5920. [PMID: 34885030 PMCID: PMC8656854 DOI: 10.3390/cancers13235920] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/22/2021] [Accepted: 11/22/2021] [Indexed: 01/23/2023] Open
Abstract
Given its pre-eminent role in the context of tumor cell growth as well as metastasis, the C-X-C motif chemokine receptor 4 (CXCR4) has attracted a lot of interest in the field of nuclear oncology, and clinical evidence on the high potential of CXCR4-targeted theranostics is constantly accumulating. Additionally, since CXCR4 also represents a key player in the orchestration of inflammatory responses to inflammatory stimuli, based on its expression on a variety of pro- and anti-inflammatory immune cells (e.g., macrophages and T-cells), CXCR4-targeted inflammation imaging has recently gained considerable attention. Therefore, after briefly summarizing the current clinical status quo of CXCR4-targeted theranostics in cancer, this review primarily focuses on imaging of a broad spectrum of inflammatory diseases via the quantification of tissue infiltration with CXCR4-expressing immune cells. An up-to-date overview of the ongoing preclinical and clinical efforts to visualize inflammation and its resolution over time is provided, and the predictive value of the CXCR4-associated imaging signal for disease outcome is discussed. Since the sensitivity and specificity of CXCR4-targeted immune cell imaging greatly relies on the availability of suitable, tailored imaging probes, recent developments in the field of CXCR4-targeted imaging agents for various applications are also addressed.
Collapse
Affiliation(s)
- Margret Schottelius
- Translational Radiopharmaceutical Sciences, Department of Nuclear Medicine and of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne (UNIL), 1011 Lausanne, Switzerland
| | - Ken Herrmann
- Department of Nuclear Medicine, German Cancer Consortium (DKTK)-University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany;
| | - Constantin Lapa
- Nuclear Medicine, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany
| |
Collapse
|
22
|
Boughdad S, Latifyan S, Fenwick C, Bouchaab H, Suffiotti M, Moslehi JJ, Salem JE, Schaefer N, Nicod-Lalonde M, Costes J, Perreau M, Michielin O, Peters S, Prior JO, Obeid M. 68Ga-DOTATOC PET/CT to detect immune checkpoint inhibitor-related myocarditis. J Immunother Cancer 2021; 9:jitc-2021-003594. [PMID: 34686542 PMCID: PMC8543755 DOI: 10.1136/jitc-2021-003594] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2021] [Indexed: 12/15/2022] Open
Abstract
Background Immune checkpoint inhibitor (ICI)-related myocarditis is a rare but potentially fatal adverse event that can occur following ICI exposure. Early diagnosis and treatment are key to improve patient outcomes. Somatostatin receptor-based positron emission tomography–CT (PET/CT) showed promising results for the assessment of myocardial inflammation, yet information regarding its value for the diagnosis of ICI-related myocarditis, especially at the early stage, is limited. Thus, we investigated the value of 68Ga-DOTA(0)-Phe(1)-Tyr(3)-octreotide (68Ga-DOTATOC) PET/CT for the early detection and diagnosis of ICI-related myocarditis. Methods Consecutive patients with clinically suspected ICI-related myocarditis from July 2018 to February 2021 were retrospectively evaluated in this single-center study. All patients underwent imaging for the detection of ICI-related myocarditis using either cardiac magnetic resonance (CMR) imaging or 68Ga-DOTATOC PET/CT. PET/CT images were acquired 90 min after the injection of 2 MBq/kg 68Ga-DOTATOC with pathological myocardial uptake in the left ventricle (LV) suggestive of myocarditis defined using a myocardium-to-background ratio of peak standard uptake value to mean intracavitary LV standard uptake (MBRpeak) value above 1.6. Patients had a full cardiological work-up including ECG, echocardiography, serum cardiac troponin I (cTnI), cardiac troponin T and creatine kinase (CK), CK-MB. Endomyocardial biopsy and inflammatory cytokine markers were also analyzed. The detection rate of ICI-related myocarditis using 68Ga-DOTATOC PET/CT and CMR was assessed. Results A total of 11 patients had clinically suspected ICI-related myocarditis; 9 underwent 68Ga -DOTATOC PET/CT. All nine (100%) patients with 68Ga-DOTATOC PET/CT presented with pathological myocardial uptake in the LV that was suggestive of myocarditis (MBRpeak of 3.2±0.8, range 2.2–4.4). Eight patients had CMR imaging and 3/8 (38%) patients had lesions evocative of myocarditis. All PET-positive patients were previously treated with a high dose of steroids and intravenous immunoglobulin prior to PET/CT had elevated serum cTnI except for one patient for whom PET/CT was delayed several days. Interestingly, in 5/6 (83%) patients who presented with concomitant myositis, pathological uptake was seen on whole-body 68Ga-DOTATOC PET/CT images in the skeletal muscles, suggesting an additional advantage of this method to assess the full extent of the disease. In contrast, four patients with CMR imaging had negative findings despite having elevated serum cTnI levels (range 20.5–5896.1 ng/mL), thus defining possible myocarditis. Newly identified immune correlates could provide specific biomarkers for the diagnosis of ICI-related myocarditis. Most tested patients (six of seven patients) had serum increases in the inflammatory cytokine interleukin (IL)-6 and in the chemokines CXCL9, CXCL10, and CXCL13, and the mass cytometry phenotypes of immune cell populations in the blood also showed correlations with myocardial inflammation. Four of five patients with myocarditis exhibited a Th1/Th2 imbalance favoring a pronounced inflammatory Th1, Th1/Th17, and Th17 CD4 memory T-cell response. The high proportion of non-classical monocytes and significantly reduced levels of CD31 in four to five patients was also consistent with an inflammatory disease. Conclusion The use of 68Ga-DOTATOC PET/CT along with immune correlates is a highly sensitive method to detect ICI-related myocarditis especially in the early stage of myocardial inflammation, as patients with elevated cTnI may present normal CMR imaging results. 68Ga-DOTATOC PET/CT is also useful for detecting concomitant myositis. These results need to be confirmed in a larger population of patients and validated against a histological gold standard if available.
Collapse
Affiliation(s)
- Sarah Boughdad
- Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
| | - Sofiya Latifyan
- Department of Medical Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - Craig Fenwick
- Immunology and allergy division, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Hasna Bouchaab
- Department of Medical Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - Madeleine Suffiotti
- Immunology and allergy division, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Javid J Moslehi
- Section of Cardio-Oncology & Immunology, Division of Cardiology and the Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, USA
| | - Joe-Elie Salem
- Cardio-Oncology Program, Pitié-Salpétrière Hospital, Paris, France, AP-HP, Pitié-Salpétrière Hospital,Sorbonne, INSERM, CIC-1901, Paris, France, Paris, France, France
| | - Niklaus Schaefer
- Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
| | - Marie Nicod-Lalonde
- Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
| | - Julien Costes
- Radiopharmacy Unit, Department of Pharmacy, Lausanne University Hospital, Lausanne, Switzerland
| | - Matthieu Perreau
- Immunology and allergy division, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Olivier Michielin
- Department of Medical Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - Solange Peters
- Department of Medical Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - John O Prior
- Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
| | - Michel Obeid
- Immunology and allergy division, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
23
|
Riou L, Toczek J, Broisat A, Ghezzi C, Djaileb L. Identifying the leukocyte uptake pattern of inflammation imaging agents: Current limitations and potential impact. J Nucl Cardiol 2021; 28:1646-1648. [PMID: 31823330 DOI: 10.1007/s12350-019-01979-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 11/22/2019] [Indexed: 10/25/2022]
Affiliation(s)
- Laurent Riou
- Laboratoire Radiopharmaceutiques Biocliniques, Faculté de Médecine de Grenoble, UMR UGA - INSERM U1039, Grenoble, France.
| | - Jakub Toczek
- Cardiovascular Molecular Imaging Laboratory, Section of Cardiovascular Medicine and Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, 06511, USA
| | - Alexis Broisat
- Laboratoire Radiopharmaceutiques Biocliniques, Faculté de Médecine de Grenoble, UMR UGA - INSERM U1039, Grenoble, France
| | - Catherine Ghezzi
- Laboratoire Radiopharmaceutiques Biocliniques, Faculté de Médecine de Grenoble, UMR UGA - INSERM U1039, Grenoble, France
| | - Loïc Djaileb
- Laboratoire Radiopharmaceutiques Biocliniques, Faculté de Médecine de Grenoble, UMR UGA - INSERM U1039, Grenoble, France
- Nuclear Medicine Department, Grenoble Alpes University Hospital, Grenoble, France
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW Current therapeutic strategies to mitigate heart failure progression after myocardial infarction involve support of endogenous repair through molecular targets. The capacity for repair varies greatly between individuals. In this review, we will assess how cardiac PET/CT enables precise characterization of early pathogenetic processes which govern ventricle remodeling and progression to heart failure. RECENT FINDINGS Inflammation in the first days after myocardial infarction predicts subsequent functional decline and can influence therapy decisions. The expansion of anti-inflammatory approaches to improve outcomes after myocardial infarction may benefit from noninvasive characterization using imaging. Novel probes also allow visualization of fibroblast transdifferentiation and activation, as a precursor to ventricle remodeling. The expanding arsenal of molecular imaging agents in parallel with new treatment options provides opportunity to harmonize diagnostic imaging with precision therapy.
Collapse
Affiliation(s)
- James T Thackeray
- Department of Nuclear Medicine, Hannover Medical School, Neuberg-Str. 1, D-30625, Hannover, Germany.
| |
Collapse
|
25
|
Sherzay R, Witte T, Derlin T, Hoepfner M, Bengel FM. Vessel Wall Inflammatory Activity as Determined by F-18 Fluorodeoxyglucose PET in Large Vessel Vasculitis Is Attenuated by Immunomodulatory Drugs. Diagnostics (Basel) 2021; 11:diagnostics11071132. [PMID: 34206366 PMCID: PMC8303651 DOI: 10.3390/diagnostics11071132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 12/12/2022] Open
Abstract
F-18 fluorodeoxyglucose (F-18 FDG) PET/CT plays an increasing role in the diagnostic workup of large vessel vasculitis (LVV); however, information on the relationship between immunosuppressive drugs and vessel wall uptake is limited. In 94 patients with a confirmed diagnosis of LVV, the vessel wall-to-liver ratio (VLR) was assessed in eight vessel segments. Patients were grouped according to intake of immunomodulatory drugs (Group 1, prednisone; Group 2, prednisone + methotrexate; and Group 3, prednisone + others) and compared to treatment-naïve individuals. A total of 54/94 (57.4%) were treated with immunomodulatory drugs (Group 1, 29/49 (53.7%); Group 2, 9/54 (16.7%); Group 3, 11/54 (20.4%); and Group 4, 5/54 (9.3%)), whereas the remainder received no therapy (40/94 (42.6%)). The mean VLR of the arterial segments correlated significantly with the hematopoietic organs (r ≥ 0.22, p ≤ 0.05), c-reactive protein (r ≥ 0.25, p ≤ 0.05), and prednisone dosage (r ≥ −0.4, p ≤ 0.05). Relative to treatment-naïve patients, a significantly lower VLR was recorded in 5/8 (62.5%) of the investigated vessel segments in Group 1 (p ≤ 0.02), in 6/8 of the vessel segments in Group 2 (75.0%, p ≤ 0.006), and in 7/8 of the segments in Group 3 (87.5%, p ≤ 0.05). In LVV, the F-18 FDG uptake in vessel wall as a marker of inflammatory activity was attenuated by immunomodulatory drugs, which provides a foundation for future serial monitoring of treatment efficacy.
Collapse
Affiliation(s)
- Romilda Sherzay
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany; (R.S.); (T.D.)
| | - Torsten Witte
- Department of Rheumatology and Immunology, Hannover Medical School, 30625 Hannover, Germany; (T.W.); (M.H.)
| | - Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany; (R.S.); (T.D.)
| | - Marius Hoepfner
- Department of Rheumatology and Immunology, Hannover Medical School, 30625 Hannover, Germany; (T.W.); (M.H.)
| | - Frank M. Bengel
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany; (R.S.); (T.D.)
- Correspondence: ; Tel.: +49-(0)-511-532-2577
| |
Collapse
|
26
|
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
|
27
|
Meester EJ, de Blois E, Krenning BJ, van der Steen AFW, Norenberg JP, van Gaalen K, Bernsen MR, de Jong M, van der Heiden K. Autoradiographical assessment of inflammation-targeting radioligands for atherosclerosis imaging: potential for plaque phenotype identification. EJNMMI Res 2021; 11:27. [PMID: 33730311 PMCID: PMC7969682 DOI: 10.1186/s13550-021-00772-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 03/05/2021] [Indexed: 12/26/2022] Open
Abstract
PURPOSE Many radioligands have been developed for the visualization of atherosclerosis by targeting inflammation. However, interpretation of in vivo signals is often limited to plaque identification. We evaluated binding of some promising radioligands in an in vitro approach in atherosclerotic plaques with different phenotypes. METHODS Tissue sections of carotid endarterectomy tissue were characterized as early plaque, fibro-calcific plaque, or phenotypically vulnerable plaque. In vitro binding assays for the radioligands [111In]In-DOTATATE; [111In]In-DOTA-JR11; [67Ga]Ga-Pentixafor; [111In]In-DANBIRT; and [111In]In-EC0800 were conducted, the expression of the radioligand targets was assessed via immunohistochemistry. Radioligand binding and expression of radioligand targets was investigated and compared. RESULTS In sections characterized as vulnerable plaque, binding was highest for [111In]In-EC0800; followed by [111In]In-DANBIRT; [67Ga]Ga-Pentixafor; [111In]In-DOTA-JR11; and [111In]In-DOTATATE (0.064 ± 0.036; 0.052 ± 0.029; 0.011 ± 0.003; 0.0066 ± 0.0021; 0.00064 ± 0.00014 %Added activity/mm2, respectively). Binding of [111In]In-DANBIRT and [111In]In-EC0800 was highest across plaque phenotypes, binding of [111In]In-DOTA-JR11 and [67Ga]Ga-Pentixafor differed most between plaque phenotypes. Binding of [111In]In-DOTATATE was the lowest across plaque phenotypes. The areas positive for cells expressing the radioligand's target differed between plaque phenotypes for all targets, with lowest percentage area of expression in early plaque sections and highest in phenotypically vulnerable plaque sections. CONCLUSIONS Radioligands targeting inflammatory cell markers showed different levels of binding in atherosclerotic plaques and among plaque phenotypes. Different radioligands might be used for plaque detection and discerning early from vulnerable plaque. [111In]In-EC0800 and [111In]In-DANBIRT appear most suitable for plaque detection, while [67Ga]Ga-Pentixafor and [111In]In-DOTA-JR11 might be best suited for differentiation between plaque phenotypes.
Collapse
Affiliation(s)
- Eric J Meester
- Department of Biomedical Engineering, Thorax Center, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Erik de Blois
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | | | - Antonius F W van der Steen
- Department of Biomedical Engineering, Thorax Center, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Jeff P Norenberg
- Radiopharmaceutical Sciences, University of New Mexico, Albuquerque, NM, USA
| | - Kim van Gaalen
- Department of Biomedical Engineering, Thorax Center, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Monique R Bernsen
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Marion de Jong
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Kim van der Heiden
- Department of Biomedical Engineering, Thorax Center, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.
| |
Collapse
|
28
|
Glasenapp A, Hess A, Thackeray JT. Molecular imaging in nuclear cardiology: Pathways to individual precision medicine. J Nucl Cardiol 2020; 27:2195-2201. [PMID: 32893320 PMCID: PMC7749093 DOI: 10.1007/s12350-020-02319-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 07/29/2020] [Indexed: 01/08/2023]
Abstract
Growth of molecular imaging bears potential to transform nuclear cardiology from a primarily diagnostic method to a precision medicine tool. Molecular targets amenable for imaging and therapeutic intervention are particularly promising to facilitate risk stratification, patient selection and exquisite guidance of novel therapies, and interrogation of systems-based interorgan communication. Non-invasive visualization of pathobiology provides valuable insights into the progression of disease and response to treatment. Specifically, inflammation, fibrosis, and neurohormonal signaling, central to the progression of cardiovascular disease and emerging therapeutic strategies, have been investigated by molecular imaging. As the number of radioligands grows, careful investigation of the binding properties and added-value of imaging should be prioritized to identify high-potential probes and facilitate translation to clinical applications. In this review, we discuss the current state of molecular imaging in cardiovascular medicine, and the challenges and opportunities ahead for cardiovascular molecular imaging to navigate the path from diagnosis to prognosis to personalized medicine.
Collapse
Affiliation(s)
- A Glasenapp
- Department of Nuclear Medicine, Hannover Medical School, Translational Cardiovascular Molecular Imaging, Carl Neuberg Str 1, 30625, Hannover, Germany
| | - A Hess
- Department of Nuclear Medicine, Hannover Medical School, Translational Cardiovascular Molecular Imaging, Carl Neuberg Str 1, 30625, Hannover, Germany
| | - J T Thackeray
- Department of Nuclear Medicine, Hannover Medical School, Translational Cardiovascular Molecular Imaging, Carl Neuberg Str 1, 30625, Hannover, Germany.
| |
Collapse
|
29
|
Wilk B, Wisenberg G, Dharmakumar R, Thiessen JD, Goldhawk DE, Prato FS. Hybrid PET/MR imaging in myocardial inflammation post-myocardial infarction. J Nucl Cardiol 2020; 27:2083-2099. [PMID: 31797321 PMCID: PMC7391987 DOI: 10.1007/s12350-019-01973-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/13/2019] [Accepted: 11/14/2019] [Indexed: 01/24/2023]
Abstract
Hybrid PET/MR imaging is an emerging imaging modality combining positron emission tomography (PET) and magnetic resonance imaging (MRI) in the same system. Since the introduction of clinical PET/MRI in 2011, it has had some impact (e.g., imaging the components of inflammation in myocardial infarction), but its role could be much greater. Many opportunities remain unexplored and will be highlighted in this review. The inflammatory process post-myocardial infarction has many facets at a cellular level which may affect the outcome of the patient, specifically the effects on adverse left ventricular remodeling, and ultimately prognosis. The goal of inflammation imaging is to track the process non-invasively and quantitatively to determine the best therapeutic options for intervention and to monitor those therapies. While PET and MRI, acquired separately, can image aspects of inflammation, hybrid PET/MRI has the potential to advance imaging of myocardial inflammation. This review contains a description of hybrid PET/MRI, its application to inflammation imaging in myocardial infarction and the challenges, constraints, and opportunities in designing data collection protocols. Finally, this review explores opportunities in PET/MRI: improved registration, partial volume correction, machine learning, new approaches in the development of PET and MRI pulse sequences, and the use of novel injection strategies.
Collapse
Affiliation(s)
- B Wilk
- Department of Medical Imaging, Western University, London, Canada.
- Lawson Health Research Institute, London, Canada.
- Collaborative Graduate Program in Molecular Imaging, Western University, London, Canada.
| | - G Wisenberg
- Department of Medical Imaging, Western University, London, Canada
- MyHealth Centre, Arva, Canada
| | - R Dharmakumar
- Biomedical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - J D Thiessen
- Department of Medical Imaging, Western University, London, Canada
- Lawson Health Research Institute, London, Canada
- Collaborative Graduate Program in Molecular Imaging, Western University, London, Canada
| | - D E Goldhawk
- Department of Medical Imaging, Western University, London, Canada
- Lawson Health Research Institute, London, Canada
- Collaborative Graduate Program in Molecular Imaging, Western University, London, Canada
| | - F S Prato
- Department of Medical Imaging, Western University, London, Canada
- Lawson Health Research Institute, London, Canada
- Collaborative Graduate Program in Molecular Imaging, Western University, London, Canada
| |
Collapse
|
30
|
Ćorović A, Wall C, Mason JC, Rudd JHF, Tarkin JM. Novel Positron Emission Tomography Tracers for Imaging Vascular Inflammation. Curr Cardiol Rep 2020; 22:119. [PMID: 32772188 PMCID: PMC7415747 DOI: 10.1007/s11886-020-01372-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Purpose of Review To provide a focused update on recent advances in positron emission tomography (PET) imaging in vascular inflammatory diseases and consider future directions in the field. Recent Findings While PET imaging with 18F-fluorodeoxyglucose (FDG) can provide a useful marker of disease activity in several vascular inflammatory diseases, including atherosclerosis and large-vessel vasculitis, this tracer lacks inflammatory cell specificity and is not a practical solution for imaging the coronary vasculature because of avid background myocardial signal. To overcome these limitations, research is ongoing to identify novel PET tracers that can more accurately track individual components of vascular immune responses. Use of these novel PET tracers could lead to a better understanding of underlying disease mechanisms and help inform the identification and stratification of patients for newly emerging immune-modulatory therapies. Summary Future research is needed to realise the true clinical translational value of PET imaging in vascular inflammatory diseases.
Collapse
Affiliation(s)
- Andrej Ćorović
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Christopher Wall
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Justin C Mason
- Cardiovascular Division, National Heart & Lung Institute, Imperial College London, London, 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. .,Cardiovascular Division, National Heart & Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
31
|
Bengel FM, Hermanns N, Thackeray JT. Radionuclide Imaging of the Molecular Mechanisms Linking Heart and Brain in Ischemic Syndromes. Circ Cardiovasc Imaging 2020; 13:e011303. [DOI: 10.1161/circimaging.120.011303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
For the heart and the brain, clinical observations suggest that an acute ischemic event experienced by one organ is associated with an increased risk for future acute events and chronic dysfunction of the reciprocal organ. Beyond atherosclerosis as a common systemic disease, various molecular mechanisms are thought to be involved in this interaction. Molecular-targeted nuclear imaging may identify the contribution of factors, such as the neurohumoral, circulatory, or especially the immune system, by combining specific radiotracers with whole-body acquisition and global as well as regional multiorgan analysis. This may be integrated with complementary functional imaging markers and systemic biomarkers for comprehensive network interrogation. Such systems-based strategies go beyond the traditional organ-centered approach and provide novel mechanistic insights, information about temporal dynamics, and a foundation for future interventions aiming at optimal preservation of function of both organs.
Collapse
Affiliation(s)
- Frank M. Bengel
- Department of Nuclear Medicine, Hannover Medical School, Germany
| | - Nele Hermanns
- Department of Nuclear Medicine, Hannover Medical School, Germany
| | | |
Collapse
|
32
|
Borchert T, Hess A, Lukačević M, Ross TL, Bengel FM, Thackeray JT. Angiotensin-converting enzyme inhibitor treatment early after myocardial infarction attenuates acute cardiac and neuroinflammation without effect on chronic neuroinflammation. Eur J Nucl Med Mol Imaging 2020; 47:1757-1768. [PMID: 32125488 PMCID: PMC7248052 DOI: 10.1007/s00259-020-04736-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 02/20/2020] [Indexed: 02/07/2023]
Abstract
Purpose Myocardial infarction (MI) triggers a local inflammatory response which orchestrates cardiac repair and contributes to concurrent neuroinflammation. Angiotensin-converting enzyme (ACE) inhibitor therapy not only attenuates cardiac remodeling by interfering with the neurohumoral system, but also influences acute leukocyte mobilization from hematopoietic reservoirs. Here, we seek to dissect the anti-inflammatory and anti-remodeling contributions of ACE inhibitors to the benefit of heart and brain outcomes after MI. Methods C57BL/6 mice underwent permanent coronary artery ligation (n = 41) or sham surgery (n = 9). Subgroups received ACE inhibitor enalapril (20 mg/kg, oral) either early (anti-inflammatory strategy; 10 days treatment beginning 3 days prior to surgery; n = 9) or delayed (anti-remodeling; continuous from 7 days post-MI; n = 16), or no therapy (n = 16). Cardiac and neuroinflammation were serially investigated using whole-body macrophage- and microglia-targeted translocator protein (TSPO) PET at 3 days, 7 days, and 8 weeks. In vivo PET signal was validated by autoradiography and histopathology. Results Myocardial infarction evoked higher TSPO signal in the infarct region at 3 days and 7 days compared with sham (p < 0.001), with concurrent elevation in brain TSPO signal (+ 18%, p = 0.005). At 8 weeks after MI, remote myocardium TSPO signal was increased, consistent with mitochondrial stress, and corresponding to recurrent neuroinflammation. Early enalapril treatment lowered the acute TSPO signal in the heart and brain by 55% (p < 0.001) and 14% (p = 0.045), respectively. The acute infarct signal predicted late functional outcome (r = 0.418, p = 0.038). Delayed enalapril treatment reduced chronic myocardial TSPO signal, consistent with alleviated mitochondrial stress. Early enalapril therapy tended to lower TSPO signal in the failing myocardium at 8 weeks after MI (p = 0.090) without an effect on chronic neuroinflammation. Conclusions Whole-body TSPO PET identifies myocardial macrophage infiltration and neuroinflammation after MI, and altered cardiomyocyte mitochondrial density in chronic heart failure. Improved chronic cardiac outcome by enalapril treatment derives partially from acute anti-inflammatory activity with complementary benefits in later stages. Whereas early ACE inhibitor therapy lowers acute neuroinflammation, chronic alleviation is not achieved by early or delayed ACE inhibitor therapy, suggesting a more complex mechanism underlying recurrent neuroinflammation in ischemic heart failure. Electronic supplementary material The online version of this article (10.1007/s00259-020-04736-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tobias Borchert
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, D-30625, Hannover, Germany
| | - Annika Hess
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, D-30625, Hannover, Germany
| | - Mario Lukačević
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, D-30625, Hannover, Germany
| | - Tobias L Ross
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, D-30625, Hannover, Germany
| | - Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, D-30625, Hannover, Germany
| | - James T Thackeray
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, D-30625, Hannover, Germany.
| |
Collapse
|