1
|
Yin G, Ruan Q, Jiang Y, Feng J, Han P, Wang Q, Li Z, Zhang J. Novel 99mTc-Labeled Mannose Derivative as a Highly Promising Single Photon Emission Computed Tomography Probe for Tumor Imaging. J Med Chem 2024; 67:15796-15806. [PMID: 39058751 DOI: 10.1021/acs.jmedchem.4c01425] [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: 07/28/2024]
Abstract
18F-2-fluoro-2-deoxy-d-glucose ([18F]FDG) has been the most used positron emission tomography imaging agent for clinical applications. Single photon emission computed tomography (SPECT) imaging is cheaper and used more widely for diagnostic use, but there is no SPECT tumor imaging agent for clinical applications comparable to [18F]FDG. Mannose is a C2 epimer of glucose and can also be transported into tumor cells via glucose transporters (GLUTs). To develop a novel SPECT tumor imaging agent with satisfactory tumor uptake and tumor/nontarget ratios, here a mannose derivative (CN7DM) was synthesized and radiolabeled with technetium-99m to prepare [99mTc]Tc-CN7DM. The six-coordinated structure of [99mTc]Tc-CN7DM was confirmed by the corresponding rhenium compound (Re-CN7DM). [99mTc]Tc-CN7DM was transported into cancer cells via GLUTs and may be trapped in the cancer cells by electrostatic attraction. The probe exhibited high uptake in tumors and low uptake in nontarget tissues in mice bearing different tumors, indicating that [99mTc]Tc-CN7DM exhibited promising potential for SPECT tumor imaging and warranted further clinical investigation.
Collapse
Affiliation(s)
- Guangxing Yin
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA (National Medical Products Administration) Key Laboratory for Research and Evaluation of Radiopharmaceuticals, College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Qing Ruan
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA (National Medical Products Administration) Key Laboratory for Research and Evaluation of Radiopharmaceuticals, College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
- Key Laboratory of Beam Technology of the Ministry of Education, School of Physics and Astronomy, Beijing Normal University, Beijing 100875, China
| | - Yuhao Jiang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA (National Medical Products Administration) Key Laboratory for Research and Evaluation of Radiopharmaceuticals, College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Junhong Feng
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA (National Medical Products Administration) Key Laboratory for Research and Evaluation of Radiopharmaceuticals, College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
- Department of Nuclear Technology and Application, China Institute of Atomic Energy, Beijing 102413, China
| | - Peiwen Han
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA (National Medical Products Administration) Key Laboratory for Research and Evaluation of Radiopharmaceuticals, College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Qianna Wang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA (National Medical Products Administration) Key Laboratory for Research and Evaluation of Radiopharmaceuticals, College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Zuojie Li
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA (National Medical Products Administration) Key Laboratory for Research and Evaluation of Radiopharmaceuticals, College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Junbo Zhang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA (National Medical Products Administration) Key Laboratory for Research and Evaluation of Radiopharmaceuticals, College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| |
Collapse
|
2
|
Maier A, Teunissen AJP, Nauta SA, Lutgens E, Fayad ZA, van Leent MMT. Uncovering atherosclerotic cardiovascular disease by PET imaging. Nat Rev Cardiol 2024; 21:632-651. [PMID: 38575752 PMCID: PMC11324396 DOI: 10.1038/s41569-024-01009-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/04/2024] [Indexed: 04/06/2024]
Abstract
Assessing atherosclerosis severity is essential for precise patient stratification. Specifically, there is a need to identify patients with residual inflammation because these patients remain at high risk of cardiovascular events despite optimal management of cardiovascular risk factors. Molecular imaging techniques, such as PET, can have an essential role in this context. PET imaging can indicate tissue-based disease status, detect early molecular changes and provide whole-body information. Advances in molecular biology and bioinformatics continue to help to decipher the complex pathogenesis of atherosclerosis and inform the development of imaging tracers. Concomitant advances in tracer synthesis methods and PET imaging technology provide future possibilities for atherosclerosis imaging. In this Review, we summarize the latest developments in PET imaging techniques and technologies for assessment of atherosclerotic cardiovascular disease and discuss the relationship between imaging readouts and transcriptomics-based plaque phenotyping.
Collapse
Affiliation(s)
- Alexander Maier
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Abraham J P Teunissen
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sheqouia A Nauta
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Esther Lutgens
- Cardiovascular Medicine and Immunology, Experimental Cardiovascular Immunology Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Zahi A Fayad
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mandy M T van Leent
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
3
|
Lee SY, Kim JH, Song JW, Min JS, Kim HJ, Kim RH, Ahn JW, Yoo H, Park K, Kim JW. Macrophage-mannose-receptor-targeted photoactivatable agent for in vivo imaging and treatment of atherosclerosis. Int J Pharm 2024; 654:123951. [PMID: 38423154 DOI: 10.1016/j.ijpharm.2024.123951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/16/2024] [Accepted: 02/25/2024] [Indexed: 03/02/2024]
Abstract
Previous studies have demonstrated the effects of theranostic agents on atherosclerotic plaques. However, there is limited information on targeted theranostics for photodynamic treatment of atherosclerosis. This study aimed to develop a macrophage-mannose-receptor-targeted photoactivatable nanoagent that regulates atherosclerosis and to evaluate its efficacy as well as safety in atherosclerotic mice. We synthesised and characterised D-mannosamine (MAN)-polyethylene glycol (PEG)-chlorin e6 (Ce6) for phototheranostic treatment of atherosclerosis. The diagnostic and therapeutic effects of MAN-PEG-Ce6 were investigated using the atherosclerotic mouse model. The hydrophobic Ce6 photosensitiser was surrounded by the hydrophilic MAN-PEG outer shell of the self-assembled nanostructure under aqueous conditions. The MAN-PEG-Ce6 was specifically internalised in macrophage-derived foam cells through receptor-mediated endocytosis. After laser irradiation, the MAN-PEG-Ce6 markedly increased singlet oxygen generation. Intravital imaging and immunohistochemistry analyses verified MAN-PEG-Ce6's specificity to plaque macrophages and its notable anti-inflammatory impact by effectively reducing mannose-receptor-positive macrophages. The toxicity assay showed that MAN-PEG-Ce6 had negligible effects on the biochemical profile and structural damage in the skin and organs. Targeted photoactivation with MAN-PEG-Ce6 thus has the potential to rapidly reduce macrophage-derived inflammatory responses in atheroma and present favourable toxicity profiles, making it a promising approach for both imaging and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Seung-Yul Lee
- Multimodal Imaging and Theranostic Laboratory, Cardiovascular Center, Korea University Guro Hospital, Seoul 08308, Republic of Korea; Division of Cardiology, Department of Internal Medicine, CHA Bundang Medical Center, Seongnam 13496, Republic of Korea
| | - Jin Hyuk Kim
- Multimodal Imaging and Theranostic Laboratory, Cardiovascular Center, Korea University Guro Hospital, Seoul 08308, Republic of Korea
| | - Joon Woo Song
- Multimodal Imaging and Theranostic Laboratory, Cardiovascular Center, Korea University Guro Hospital, Seoul 08308, Republic of Korea
| | - Ji Seon Min
- Department of Systems Biotechnology, Chung-Ang University, Anseong 17546, Republic of Korea
| | - Hyun Jung Kim
- Multimodal Imaging and Theranostic Laboratory, Cardiovascular Center, Korea University Guro Hospital, Seoul 08308, Republic of Korea
| | - Ryeong Hyun Kim
- Multimodal Imaging and Theranostic Laboratory, Cardiovascular Center, Korea University Guro Hospital, Seoul 08308, Republic of Korea
| | - Jae Won Ahn
- Department of Systems Biotechnology, Chung-Ang University, Anseong 17546, Republic of Korea
| | - Hongki Yoo
- Department of Mechanical Engineering, KAIST, Daejeon 34141, Republic of Korea.
| | - Kyeongsoon Park
- Department of Systems Biotechnology, Chung-Ang University, Anseong 17546, Republic of Korea.
| | - Jin Won Kim
- Multimodal Imaging and Theranostic Laboratory, Cardiovascular Center, Korea University Guro Hospital, Seoul 08308, Republic of Korea.
| |
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: 1.0] [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
|
Gondry O, Xavier C, Raes L, Heemskerk J, Devoogdt N, Everaert H, Breckpot K, Lecocq Q, Decoster L, Fontaine C, Schallier D, Aspeslagh S, Vaneycken I, Raes G, Van Ginderachter JA, Lahoutte T, Caveliers V, Keyaerts M. Phase I Study of [ 68Ga]Ga-Anti-CD206-sdAb for PET/CT Assessment of Protumorigenic Macrophage Presence in Solid Tumors (MMR Phase I). J Nucl Med 2023; 64:1378-1384. [PMID: 37474271 PMCID: PMC10478821 DOI: 10.2967/jnumed.122.264853] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 05/05/2023] [Indexed: 07/22/2023] Open
Abstract
Macrophages play an important role throughout the body. Antiinflammatory macrophages expressing the macrophage mannose receptor (MMR, CD206) are involved in disease development, ranging from oncology to atherosclerosis and rheumatoid arthritis. [68Ga]Ga-NOTA-anti-CD206 single-domain antibody (sdAb) is a PET tracer targeting CD206. This first-in-human study, as its primary objective, evaluated the safety, biodistribution, and dosimetry of this tracer. The secondary objective was to assess its tumor uptake. Methods: Seven patients with a solid tumor of at least 10 mm, an Eastern Cooperative Oncology Group score of 0 or 1, and good renal and hepatic function were included. Safety was evaluated using clinical examination and blood sampling before and after injection. For biodistribution and dosimetry, PET/CT was performed at 11, 90, and 150 min after injection; organs showing tracer uptake were delineated, and dosimetry was evaluated. Blood samples were obtained at selected time points for blood clearance. Metabolites in blood and urine were assessed. Results: Seven patients were injected with, on average, 191 MBq of [68Ga]Ga-NOTA-anti-CD206-sdAb. Only 1 transient adverse event of mild severity was considered to be possibly, although unlikely, related to the study drug (headache, Common Terminology Criteria for Adverse Events grade 1). The blood clearance was fast, with less than 20% of the injected activity remaining after 80 min. There was uptake in the liver, kidneys, spleen, adrenals, and red bone marrow. The average effective dose from the radiopharmaceutical was 4.2 mSv for males and 5.2 mSv for females. No metabolites were detected. Preliminary data of tumor uptake in cancer lesions showed higher uptake in the 3 patients who subsequently progressed than in the 3 patients without progression. One patient could not be evaluated because of technical failure. Conclusion: [68Ga]Ga-NOTA-anti-CD206-sdAb is safe and well tolerated. It shows rapid blood clearance and renal excretion, enabling high contrast-to-noise imaging at 90 min after injection. The radiation dose is comparable to that of routinely used PET tracers. These findings and the preliminary results in cancer patients warrant further investigation of this tracer in phase II clinical trials.
Collapse
Affiliation(s)
- Odrade Gondry
- MIMA, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium;
- Nuclear Medicine Department, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Catarina Xavier
- MIMA, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Laurens Raes
- Nuclear Medicine Department, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Johannes Heemskerk
- Nuclear Medicine Department, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Nick Devoogdt
- MIMA, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hendrik Everaert
- Nuclear Medicine Department, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Quentin Lecocq
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lore Decoster
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Christel Fontaine
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Denis Schallier
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Sandrine Aspeslagh
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Ilse Vaneycken
- Nuclear Medicine Department, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Geert Raes
- Cellular and Molecular Immunology, Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; and
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Jo A Van Ginderachter
- Cellular and Molecular Immunology, Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; and
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Tony Lahoutte
- MIMA, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
- Nuclear Medicine Department, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Vicky Caveliers
- MIMA, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
- Nuclear Medicine Department, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Marleen Keyaerts
- MIMA, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
- Nuclear Medicine Department, Universitair Ziekenhuis Brussel, Brussels, Belgium
| |
Collapse
|
6
|
Li J, Xu J, Zhang W, Li P, Zhang W, Wang H, Tang B. Detection and Imaging of Active Substances in Early Atherosclerotic Lesions Using Fluorescent Probes. Chembiochem 2023; 24:e202300105. [PMID: 36898970 DOI: 10.1002/cbic.202300105] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/12/2023]
Abstract
Atherosclerosis (AS) is a vascular disease caused by chronic inflammation and lipids that is the main cause of myocardial infarction, stroke and other cardiovascular diseases. Atherosclerosis is often difficult to detect in its early stages due to the absence of clinically significant vascular stenosis. This is not conducive to early intervention or treatment of the disease. Over the past decade, researchers have developed various imaging methods for the detection and imaging of atherosclerosis. At the same time, more and more biomarkers are being found that can be used as targets for detecting atherosclerosis. Therefore, the development of a variety of imaging methods and a variety of targeted imaging probes is an important project to achieve early assessment and treatment of atherosclerosis. This paper provides a comprehensive review of the optical probes used to detect and target atherosclerosis imaging in recent years, and describes the current challenges and future development directions.
Collapse
Affiliation(s)
- Jin Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for, Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Jiheng Xu
- School of Materials Science and Engineering, Shandong University, Jinan, 250014, P. R. China
| | - Wei Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for, Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Ping Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for, Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Wen Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for, Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Hui Wang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for, Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for, Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| |
Collapse
|
7
|
Campbell E, Jordan C, Gilmour R. Fluorinated carbohydrates for 18F-positron emission tomography (PET). Chem Soc Rev 2023; 52:3599-3626. [PMID: 37171037 PMCID: PMC10243284 DOI: 10.1039/d3cs00037k] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Indexed: 05/13/2023]
Abstract
Carbohydrate diversity is foundational in the molecular literacy that regulates cellular function and communication. Consequently, delineating and leveraging this structure-function interplay continues to be a core research objective in the development of candidates for biomedical diagnostics. A totemic example is the ubiquity of 2-deoxy-2-[18F]-fluoro-D-glucose (2-[18F]-FDG) as a radiotracer for positron emission tomography (PET), in which metabolic trapping is harnessed. Building on this clinical success, more complex sugars with unique selectivities are gaining momentum in molecular recognition and personalised medicine: this reflects the opportunities that carbohydrate-specific targeting affords in a broader sense. In this Tutorial Review, key milestones in the development of 2-[18F]-FDG and related glycan-based radiotracers for PET are described, with their diagnostic functions, to assist in navigating this rapidly expanding field of interdisciplinary research.
Collapse
Affiliation(s)
- Emma Campbell
- Organisch-Chemisches Institut, Westfälische Wilhelms-Universität Münster Corrensstraße 36, 48149, Münster, Germany.
- Cells in Motion Interfaculty Centre, Westfälische Wilhelms-Universität Münster, Röntgenstraße 16, 48149, Münster, Germany
| | - Christina Jordan
- Organisch-Chemisches Institut, Westfälische Wilhelms-Universität Münster Corrensstraße 36, 48149, Münster, Germany.
- Cells in Motion Interfaculty Centre, Westfälische Wilhelms-Universität Münster, Röntgenstraße 16, 48149, Münster, Germany
| | - Ryan Gilmour
- Organisch-Chemisches Institut, Westfälische Wilhelms-Universität Münster Corrensstraße 36, 48149, Münster, Germany.
- Cells in Motion Interfaculty Centre, Westfälische Wilhelms-Universität Münster, Röntgenstraße 16, 48149, Münster, Germany
| |
Collapse
|
8
|
Xia W, Singh N, Goel S, Shi S. Molecular Imaging of Innate Immunity and Immunotherapy. Adv Drug Deliv Rev 2023; 198:114865. [PMID: 37182699 DOI: 10.1016/j.addr.2023.114865] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/17/2023] [Accepted: 05/03/2023] [Indexed: 05/16/2023]
Abstract
The innate immune system plays a key role as the first line of defense in various human diseases including cancer, cardiovascular and inflammatory diseases. In contrast to tissue biopsies and blood biopsies, in vivo imaging of the innate immune system can provide whole body measurements of immune cell location and function and changes in response to disease progression and therapy. Rationally developed molecular imaging strategies can be used in evaluating the status and spatio-temporal distributions of the innate immune cells in near real-time, mapping the biodistribution of novel innate immunotherapies, monitoring their efficacy and potential toxicities, and eventually for stratifying patients that are likely to benefit from these immunotherapies. In this review, we will highlight the current state-of-the-art in noninvasive imaging techniques for preclinical imaging of the innate immune system particularly focusing on cell trafficking, biodistribution, as well as pharmacokinetics and dynamics of promising immunotherapies in cancer and other diseases; discuss the unmet needs and current challenges in integrating imaging modalities and immunology and suggest potential solutions to overcome these barriers.
Collapse
Affiliation(s)
- Wenxi Xia
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, United States
| | - Neetu Singh
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, United States
| | - Shreya Goel
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, United States; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, United States; Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84112, United States
| | - Sixiang Shi
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, United States; Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84112, United States.
| |
Collapse
|
9
|
Nakahara T, Strauss HW, Narula J, Jinzaki M. Vulnerable Plaque Imaging. Semin Nucl Med 2023; 53:230-240. [PMID: 36333157 DOI: 10.1053/j.semnuclmed.2022.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/27/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022]
Abstract
Atherosclerotic plaques progress as a result of inflammation. Both invasive and noninvasive imaging techniques have been developed to identify and characterize plaque as vulnerable (more likely to rupture and cause a clinical event). Imaging techniques to identify vulnerable include identifying vessels with focal subendothelial collections of I) inflammatory cells; II) lipid/ fatty acid; III) local regions of hypoxia; IV) local expression of angiogenesis factors; V) local expression of protease; VI) intravascular foci of thrombus; hemorrhage (most often seen in the aftermath of a clinical event); VII) apoptosis and VIII) microcalcification. This review provides an overview of atherosclerotic plaque progression and tracers which can visualize specific molecules associated with vulnerability.
Collapse
Affiliation(s)
- Takehiro Nakahara
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan.
| | - H William Strauss
- Division of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Jagat Narula
- Division of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Mahahiro Jinzaki
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
10
|
Ma X, Zhang MJ, Wang J, Zhang T, Xue P, Kang Y, Sun ZJ, Xu Z. Emerging Biomaterials Imaging Antitumor Immune Response. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2204034. [PMID: 35728795 DOI: 10.1002/adma.202204034] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/19/2022] [Indexed: 06/15/2023]
Abstract
Immunotherapy is one of the most promising clinical modalities for the treatment of malignant tumors and has shown excellent therapeutic outcomes in clinical settings. However, it continues to face several challenges, including long treatment cycles, high costs, immune-related adverse events, and low response rates. Thus, it is critical to predict the response rate to immunotherapy by using imaging technology in the preoperative and intraoperative. Here, the latest advances in nanosystem-based biomaterials used for predicting responses to immunotherapy via the imaging of immune cells and signaling molecules in the immune microenvironment are comprehensively summarized. Several imaging methods, such as fluorescence imaging, magnetic resonance imaging, positron emission tomography imaging, ultrasound imaging, and photoacoustic imaging, used in immune predictive imaging, are discussed to show the potential of nanosystems for distinguishing immunotherapy responders from nonresponders. Nanosystem-based biomaterials aided by various imaging technologies are expected to enable the effective prediction and diagnosis in cases of tumors, inflammation, and other public diseases.
Collapse
Affiliation(s)
- Xianbin Ma
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
- Institute of Engineering Medicine, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Meng-Jie Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| | - Jingting Wang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| | - Tian Zhang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| | - Peng Xue
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| | - Yuejun Kang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| | - Zhi-Jun Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| | - Zhigang Xu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| |
Collapse
|
11
|
Kondakov A, Berdalin A, Beregov M, Lelyuk V. Emerging Nuclear Medicine Imaging of Atherosclerotic Plaque Formation. J Imaging 2022; 8:261. [PMID: 36286355 PMCID: PMC9605050 DOI: 10.3390/jimaging8100261] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/01/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022] Open
Abstract
Atherosclerosis is a chronic widespread cardiovascular disease and a major predisposing factor for cardiovascular events, among which there are myocardial infarction and ischemic stroke. Atherosclerotic plaque formation is a process that involves different mechanisms, of which inflammation is the most common. Plenty of radiopharmaceuticals were developed to elucidate the process of plaque formation at different stages, some of which were highly specific for atherosclerotic plaque. This review summarizes the current nuclear medicine imaging landscape of preclinical and small-scale clinical studies of these specific RPs, which are not as widespread as labeled FDG, sodium fluoride, and choline. These include oxidation-specific epitope imaging, macrophage, and other cell receptors visualization, neoangiogenesis, and macrophage death imaging. It is shown that specific radiopharmaceuticals have strength in pathophysiologically sound imaging of the atherosclerotic plaques at different stages, but this also may induce problems with the signal registration for low-volume plaques in the vascular wall.
Collapse
Affiliation(s)
- Anton Kondakov
- Ultrasound and Functional Diagnostics Department, Federal Center of Brain Research and Neurotechnologies, 117513 Moscow, Russia
- Radiology and Radiotherapy Department, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Alexander Berdalin
- Ultrasound and Functional Diagnostics Department, Federal Center of Brain Research and Neurotechnologies, 117513 Moscow, Russia
| | - Mikhail Beregov
- Ultrasound and Functional Diagnostics Department, Federal Center of Brain Research and Neurotechnologies, 117513 Moscow, Russia
| | - Vladimir Lelyuk
- Ultrasound and Functional Diagnostics Department, Federal Center of Brain Research and Neurotechnologies, 117513 Moscow, Russia
| |
Collapse
|
12
|
Toribio M, Wilks MQ, Hedgire S, Lu MT, Cetlin M, Wang M, Alhallak I, Durbin CG, White KS, Wallis Z, Schnittman SR, Stanley TL, El-Fakhri G, Lee H, Autissier P, Zanni MV, Williams KC, Grinspoon SK. Increased Macrophage-Specific Arterial Infiltration Relates to Non-calcified Plaque and Systemic Immune Activation in People with HIV. J Infect Dis 2022; 226:1823-1833. [PMID: 35856671 DOI: 10.1093/infdis/jiac301] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Persistent immune activation is thought to contribute to heightened atherosclerotic cardiovascular disease (ASCVD) risk among people with HIV (PWH). METHODS Participants (≥18 years) with versus without HIV and without history of clinical ASCVD were enrolled. We hypothesized that increased macrophage-specific arterial infiltration would relate to plaque composition and systemic immune activation among PWH. We applied a novel targeted molecular imaging approach [technetium-99 m (99mTc)-tilmanocept single photon emission computed tomography (SPECT)/CT] and comprehensive immune phenotyping. RESULTS Aortic 99mTc-tilmanocept uptake was significantly higher among PWH (N = 20) versus participants without HIV (N = 10) with similar 10-year ASCVD risk (P = 0.02). Among PWH, but not among participants without HIV, non-calcified aortic plaque volume related directly to aortic 99mTc-tilmanocept uptake at different uptake thresholds. An interaction (P = 0.001) was seen between HIV status and non-calcified plaque volume, but not calcified plaque (P = 0.83). Systemic levels of caspase-1 (P = 0.004), CD14-CD16+ (non-classical/patrolling/homing) monocytes (P = 0.0004) and CD8+ T-cells (P = 0.005) related positively and CD4+/CD8 + T-cell ratio (P = 0.02) inversely to aortic 99mTc-tilmanocept uptake volume. CONCLUSIONS Macrophage-specific arterial infiltration was higher among PWH and related to non-calcified aortic plaque volume only among PWH. Key systemic markers of immune activation relating to macrophage-specific arterial infiltration may contribute to heightened ASCVD risk among PWH.
Collapse
Affiliation(s)
- Mabel Toribio
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Moses Q Wilks
- Gordon Center for Medical Imaging, Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Sandeep Hedgire
- Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Michael T Lu
- Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Madeline Cetlin
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Melissa Wang
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Iad Alhallak
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Claudia G Durbin
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Kevin S White
- Biology Department, Boston College, Chestnut Hill, MA, USA
| | - Zoey Wallis
- Biology Department, Boston College, Chestnut Hill, MA, USA
| | - Samuel R Schnittman
- Division of Infectious Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Takara L Stanley
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Georges El-Fakhri
- Gordon Center for Medical Imaging, Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Hang Lee
- Biostatistics Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Markella V Zanni
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Steven K Grinspoon
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
13
|
van der Heide CD, Dalm SU. Radionuclide imaging and therapy directed towards the tumor microenvironment: a multi-cancer approach for personalized medicine. Eur J Nucl Med Mol Imaging 2022; 49:4616-4641. [PMID: 35788730 PMCID: PMC9606105 DOI: 10.1007/s00259-022-05870-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/09/2022] [Indexed: 12/19/2022]
Abstract
Targeted radionuclide theranostics is becoming more and more prominent in clinical oncology. Currently, most nuclear medicine compounds researched for cancer theranostics are directed towards targets expressed in only a small subset of cancer types, limiting clinical applicability. The identification of cancer-specific targets that are (more) universally expressed will allow more cancer patients to benefit from these personalized nuclear medicine–based interventions. A tumor is not merely a collection of cancer cells, it also comprises supporting stromal cells embedded in an altered extracellular matrix (ECM), together forming the tumor microenvironment (TME). Since the TME is less genetically unstable than cancer cells, and TME phenotypes can be shared between cancer types, it offers targets that are more universally expressed. The TME is characterized by the presence of altered processes such as hypoxia, acidity, and increased metabolism. Next to the ECM, the TME consists of cancer-associated fibroblasts (CAFs), macrophages, endothelial cells forming the neo-vasculature, immune cells, and cancer-associated adipocytes (CAAs). Radioligands directed at the altered processes, the ECM, and the cellular components of the TME have been developed and evaluated in preclinical and clinical studies for targeted radionuclide imaging and/or therapy. In this review, we provide an overview of the TME targets and their corresponding radioligands. In addition, we discuss what developments are needed to further explore the TME as a target for radionuclide theranostics, with the hopes of stimulating the development of novel TME radioligands with multi-cancer, or in some cases even pan-cancer, application.
Collapse
Affiliation(s)
| | - Simone U Dalm
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
14
|
Fernández-García V, Boscá L. Use of 11C-acetate PET imaging in the evaluation of advanced atherogenic lesions. J Nucl Cardiol 2022; 29:1277-1279. [PMID: 33665731 DOI: 10.1007/s12350-021-02562-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 01/27/2021] [Indexed: 02/07/2023]
Abstract
The use of 11C-acetate as a PET/CT tracer for atherosclerotic lesions preferentially labels anti-inflammatory/pro-resolution intra-plaque macrophages. An overview of the mechanisms involved in the selective uptake.
Collapse
Affiliation(s)
- Victoria Fernández-García
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain.
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
| |
Collapse
|
15
|
Fernandes B, Feltes PK, Luft C, Nazario LR, Jeckel CMM, Antunes IF, Elsinga PH, de Vries EFJ. Potential PET tracers for imaging of tumor-associated macrophages. EJNMMI Radiopharm Chem 2022; 7:11. [PMID: 35526184 PMCID: PMC9081075 DOI: 10.1186/s41181-022-00163-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 04/20/2022] [Indexed: 01/05/2023] Open
Abstract
The increasing incidence of cancer over the years is one of the most challenging problems in healthcare. As cancer progresses, the recruitment of several immune cells is triggered. Infiltration of tumor-associated macrophages (TAMs) is correlated with poor patient prognosis. Since TAMs constitute a big portion of the tumor mass, targeting these cells seems to be an attractive approach for cancer immunotherapy. Additionally, TAM assessment using non-invasive imaging techniques, such as positron emission tomography (PET), might provide a better understanding of the role of TAMs in cancer, and a means for tumor profile characterization, patient selection for individualized immunotherapy and treatment monitoring. Imaging of TAMs using PET tracers is still in its infancy. TAMs have several characteristics that could be exploited as potential targets for imaging. Various PET tracers for these TAM biomarkers have been developed, although often in the context of (neuro)inflammatory diseases rather than cancer. Since macrophages in inflammatory diseases express similar biomarkers as TAMs, these PET tracers could potentially also be applied for the assessment of TAMs in the tumor microenvironment. Therefore, the present review provides an overview of the TAM biomarkers, for which potential PET tracers are available and discusses the status of these tracers.
Collapse
Affiliation(s)
- Bruna Fernandes
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, P.O. Box 30 001, 9700 RB, Groningen, The Netherlands.,Graduate Program in Biomedical Gerontology, School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Paula Kopschina Feltes
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, P.O. Box 30 001, 9700 RB, Groningen, The Netherlands
| | - Carolina Luft
- Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,Laboratory of Pediatric Physical Activity, Infant Center, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Luiza Reali Nazario
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, P.O. Box 30 001, 9700 RB, Groningen, The Netherlands
| | - Cristina Maria Moriguchi Jeckel
- Graduate Program in Biomedical Gerontology, School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Ines F Antunes
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, P.O. Box 30 001, 9700 RB, Groningen, The Netherlands
| | - Philip H Elsinga
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, P.O. Box 30 001, 9700 RB, Groningen, The Netherlands
| | - Erik F J de Vries
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, P.O. Box 30 001, 9700 RB, Groningen, The Netherlands.
| |
Collapse
|
16
|
Crișan G, Moldovean-Cioroianu NS, Timaru DG, Andrieș G, Căinap C, Chiș V. Radiopharmaceuticals for PET and SPECT Imaging: A Literature Review over the Last Decade. Int J Mol Sci 2022; 23:5023. [PMID: 35563414 PMCID: PMC9103893 DOI: 10.3390/ijms23095023] [Citation(s) in RCA: 82] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/23/2022] [Accepted: 04/28/2022] [Indexed: 02/04/2023] Open
Abstract
Positron emission tomography (PET) uses radioactive tracers and enables the functional imaging of several metabolic processes, blood flow measurements, regional chemical composition, and/or chemical absorption. Depending on the targeted processes within the living organism, different tracers are used for various medical conditions, such as cancer, particular brain pathologies, cardiac events, and bone lesions, where the most commonly used tracers are radiolabeled with 18F (e.g., [18F]-FDG and NA [18F]). Oxygen-15 isotope is mostly involved in blood flow measurements, whereas a wide array of 11C-based compounds have also been developed for neuronal disorders according to the affected neuroreceptors, prostate cancer, and lung carcinomas. In contrast, the single-photon emission computed tomography (SPECT) technique uses gamma-emitting radioisotopes and can be used to diagnose strokes, seizures, bone illnesses, and infections by gauging the blood flow and radio distribution within tissues and organs. The radioisotopes typically used in SPECT imaging are iodine-123, technetium-99m, xenon-133, thallium-201, and indium-111. This systematic review article aims to clarify and disseminate the available scientific literature focused on PET/SPECT radiotracers and to provide an overview of the conducted research within the past decade, with an additional focus on the novel radiopharmaceuticals developed for medical imaging.
Collapse
Affiliation(s)
- George Crișan
- Faculty of Physics, Babeş-Bolyai University, Str. M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania; (G.C.); (N.S.M.-C.); (D.-G.T.)
- Department of Nuclear Medicine, County Clinical Hospital, Clinicilor 3-5, 400006 Cluj-Napoca, Romania;
| | | | - Diana-Gabriela Timaru
- Faculty of Physics, Babeş-Bolyai University, Str. M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania; (G.C.); (N.S.M.-C.); (D.-G.T.)
| | - Gabriel Andrieș
- Department of Nuclear Medicine, County Clinical Hospital, Clinicilor 3-5, 400006 Cluj-Napoca, Romania;
| | - Călin Căinap
- The Oncology Institute “Prof. Dr. Ion Chiricuţă”, Republicii 34-36, 400015 Cluj-Napoca, Romania;
| | - Vasile Chiș
- Faculty of Physics, Babeş-Bolyai University, Str. M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania; (G.C.); (N.S.M.-C.); (D.-G.T.)
- Institute for Research, Development and Innovation in Applied Natural Sciences, Babeș-Bolyai University, Str. Fântânele 30, 400327 Cluj-Napoca, Romania
| |
Collapse
|
17
|
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
|
18
|
Positron Emission Tomography in Atherosclerosis Research. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2419:825-839. [PMID: 35238004 DOI: 10.1007/978-1-0716-1924-7_50] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Positron emission tomography (PET) is a quantitative imaging technique that uses molecules labeled with positron-emitting radionuclides to visualize and measure biochemical processes in the tissues of living subjects. In recent years, different PET tracers have been evaluated for their ability to characterize the atherosclerotic process in order to study the activity of the disease. Here, we describe detailed PET methods for preclinical studies of atherosclerosis and summarize the key methodological aspects of PET imaging in clinical studies of atherosclerosis.
Collapse
|
19
|
Li XG, Velikyan I, Viitanen R, Roivainen A. PET radiopharmaceuticals for imaging inflammatory diseases. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00075-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
20
|
Thanh Nguyen TD, Marasini R, Aryal S. Re-engineered imaging agent using biomimetic approaches. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1762. [PMID: 34698438 PMCID: PMC8758533 DOI: 10.1002/wnan.1762] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/25/2021] [Indexed: 01/03/2023]
Abstract
Recent progress in biomedical technology, the clinical bioimaging, has a greater impact on the diagnosis, treatment, and prevention of disease, especially by early intervention and precise therapy. Varieties of organic and inorganic materials either in the form of small molecules or nano-sized materials have been engineered as a contrast agent (CA) to enhance image resolution among different tissues for the detection of abnormalities such as cancer and vascular occlusion. Among different innovative imaging agents, contrast agents coupled with biologically derived endogenous platform shows the promising application in the biomedical field, including drug delivery and bioimaging. Strategy using biocomponents such as cells or products of cells as a delivery system predominantly reduces the toxic behavior of its cargo, as these systems reduce non-specific distribution by navigating its cargo toward the targeted location. The hypothesis is that depending on the original biological role of the naïve cell, the contrast agents carried by such a system can provide corresponding natural designated behavior. Therefore, by combining properties of conventional synthetic molecules and nanomaterials with endogenous cell body, new solutions in the field of bioimaging to overcome biological barriers have been offered as innovative bioengineering. In this review, we will discuss the engineering of cell and cell-derived components as a delivery system for various contrast agents to achieve clinically relevant contrast for diagnosis and study underlining mechanism of disease progression. This article is categorized under: Nanotechnology Approaches to Biology > Cells at the Nanoscale Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Tuyen Duong Thanh Nguyen
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Ramesh Marasini
- Department of Chemistry, Nanotechnology Innovation Center of Kansas State, Kansas State Univeristy, Manhattan, KS
| | - Santosh Aryal
- Department of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler, Tyler, Texas 75799, USA
| |
Collapse
|
21
|
Functionally Heterogenous Macrophage Subsets in the Pathogenesis of Giant Cell Arteritis: Novel Targets for Disease Monitoring and Treatment. J Clin Med 2021; 10:jcm10214958. [PMID: 34768479 PMCID: PMC8585092 DOI: 10.3390/jcm10214958] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/20/2021] [Accepted: 10/23/2021] [Indexed: 12/19/2022] Open
Abstract
Giant cell arteritis (GCA) is a granulomatous large-vessel vasculitis that affects adults above 50 years of age. In GCA, circulating monocytes are recruited to the inflamed arteries. With cues from the vascular microenvironment, they differentiate into macrophages and play important roles in the pathogenesis of GCA via pro-inflammatory cytokine production and vascular remodeling. However, a deeper understanding of macrophage heterogeneity in GCA pathogenesis is needed to assist the development of novel diagnostic tools and targeted therapies. Here, we review the current knowledge on macrophage heterogeneity and diverse functions of macrophage subsets in the pathogenesis of GCA. We next discuss the possibility to exploit their heterogeneity as a source of novel biomarkers and as targets for nuclear imaging. Finally, we discuss novel macrophage-targeted therapies and future directions for targeting these cells in GCA.
Collapse
|
22
|
Galiuto L, Leccisotti L, Locorotondo G, Porto I, Burzotta F, Trani C, Niccoli G, Leone AM, Danza ML, Melita V, Fedele E, Stefanelli A, Giordano A, Crea F. Coronary plaque instability assessed by positron emission tomography and optical coherence tomography. Ann Nucl Med 2021; 35:1136-1146. [PMID: 34273103 PMCID: PMC8408060 DOI: 10.1007/s12149-021-01651-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 06/27/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND Non-ST-elevation myocardial infarction (NSTEMI) and unstable angina (UA) are caused often by destabilization of non-flow limiting inflamed coronary artery plaques. 18F-fluorodeoxyglucose (FDG) uptake with positron emission tomography/computed tomography (PET/CT) reveals plaque inflammation, while intracoronary optical coherence tomography (OCT) reliably identifies morphological features of coronary instability, such as plaque rupture or erosion. We aimed to prospectively compare these two innovative biotechnologies in the characterization of coronary artery inflammation, which has never been attempted before. METHODS OCT and FDG PET/CT were performed in 18 patients with single vessel coronary artery disease, treated by percutaneous coronary intervention (PCI) with stent implantation, divided into 2 groups: NSTEMI/UA (n = 10) and stable angina (n = 8) patients. RESULTS Plaque rupture/erosion recurred more frequently [100% vs 25%, p = 0.001] and FDG uptake was greater [TBR median 1.50 vs 0.87, p = 0.004] in NSTEMI/UA than stable angina patients. FDG uptake resulted greater in patients with than without plaque rupture/erosion [1.2 (0.86-1.96) vs 0.87 (0.66-1.07), p = 0.013]. Among NSTEMI/UA patients, no significant difference in FDG uptake was found between ruptured and eroded plaques. The highest FDG uptake values were found in ruptured plaques, belonging to patients with NSTEMI/UA. OCT and PET/CT agreed in 72% of patients [p = 0.018]: 100% of patients with plaque rupture/erosion and increased FDG uptake had NSTEMI/UA. CONCLUSION For the first time, we demonstrated that the correspondence between increased FDG uptake with PET/CT and morphology of coronary plaque instability at OCT is high.
Collapse
Affiliation(s)
- L Galiuto
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168, Rome, Italy
| | - L Leccisotti
- Nuclear Medicine Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - G Locorotondo
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168, Rome, Italy.
| | - I Porto
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168, Rome, Italy
| | - F Burzotta
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168, Rome, Italy
| | - C Trani
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168, Rome, Italy
| | - G Niccoli
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168, Rome, Italy
| | - A M Leone
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168, Rome, Italy
| | - M L Danza
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168, Rome, Italy
| | - V Melita
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168, Rome, Italy
| | - E Fedele
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168, Rome, Italy
| | - A Stefanelli
- Nuclear Medicine Institute, Università Cattolica del Sacro Cuore, Rome, Italy
| | - A Giordano
- Nuclear Medicine Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Nuclear Medicine Institute, Università Cattolica del Sacro Cuore, Rome, Italy
| | - F Crea
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168, Rome, Italy
| |
Collapse
|
23
|
Balogh V, MacAskill MG, Hadoke PWF, Gray GA, Tavares AAS. Positron Emission Tomography Techniques to Measure Active Inflammation, Fibrosis and Angiogenesis: Potential for Non-invasive Imaging of Hypertensive Heart Failure. Front Cardiovasc Med 2021; 8:719031. [PMID: 34485416 PMCID: PMC8416043 DOI: 10.3389/fcvm.2021.719031] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/22/2021] [Indexed: 12/11/2022] Open
Abstract
Heart failure, which is responsible for a high number of deaths worldwide, can develop due to chronic hypertension. Heart failure can involve and progress through several different pathways, including: fibrosis, inflammation, and angiogenesis. Early and specific detection of changes in the myocardium during the transition to heart failure can be made via the use of molecular imaging techniques, including positron emission tomography (PET). Traditional cardiovascular PET techniques, such as myocardial perfusion imaging and sympathetic innervation imaging, have been established at the clinical level but are often lacking in pathway and target specificity that is important for assessment of heart failure. Therefore, there is a need to identify new PET imaging markers of inflammation, fibrosis and angiogenesis that could aid diagnosis, staging and treatment of hypertensive heart failure. This review will provide an overview of key mechanisms underlying hypertensive heart failure and will present the latest developments in PET probes for detection of cardiovascular inflammation, fibrosis and angiogenesis. Currently, selective PET probes for detection of angiogenesis remain elusive but promising PET probes for specific targeting of inflammation and fibrosis are rapidly progressing into clinical use.
Collapse
Affiliation(s)
- Viktoria Balogh
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom.,Edinburgh Imaging, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Mark G MacAskill
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom.,Edinburgh Imaging, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Patrick W F Hadoke
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Gillian A Gray
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Adriana A S Tavares
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom.,Edinburgh Imaging, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
24
|
Song JW, Nam HS, Ahn JW, Park HS, Kang DO, Kim HJ, Kim YH, Han J, Choi JY, Lee SY, Kim S, Oh WY, Yoo H, Park K, Kim JW. Macrophage targeted theranostic strategy for accurate detection and rapid stabilization of the inflamed high-risk plaque. Theranostics 2021; 11:8874-8893. [PMID: 34522216 PMCID: PMC8419038 DOI: 10.7150/thno.59759] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 07/21/2021] [Indexed: 12/17/2022] Open
Abstract
Rationale: Inflammation plays a pivotal role in the pathogenesis of the acute coronary syndrome. Detecting plaques with high inflammatory activity and specifically treating those lesions can be crucial to prevent life-threatening cardiovascular events. Methods: Here, we developed a macrophage mannose receptor (MMR)-targeted theranostic nanodrug (mannose-polyethylene glycol-glycol chitosan-deoxycholic acid-cyanine 7-lobeglitazone; MMR-Lobe-Cy) designed to identify inflammatory activity as well as to deliver peroxisome proliferator-activated gamma (PPARγ) agonist, lobeglitazone, specifically to high-risk plaques based on the high mannose receptor specificity. The MMR-Lobe-Cy was intravenously injected into balloon-injured atheromatous rabbits and serial in vivo optical coherence tomography (OCT)-near-infrared fluorescence (NIRF) structural-molecular imaging was performed. Results: One week after MMR-Lobe-Cy administration, the inflammatory NIRF signals in the plaques notably decreased compared to the baseline whereas the signals in saline controls even increased over time. In accordance with in vivo imaging findings, ex vivo NIRF signals on fluorescence reflectance imaging (FRI) and plaque inflammation by immunostainings significantly decreased compared to oral lobeglitazone group or saline controls. The anti-inflammatory effect of MMR-Lobe-Cy was mediated by inhibition of TLR4/NF-κB pathway. Furthermore, acute resolution of inflammation altered the inflamed plaque into a stable phenotype with less macrophages and collagen-rich matrix. Conclusion: Macrophage targeted PPARγ activator labeled with NIRF rapidly stabilized the inflamed plaques in coronary sized artery, which could be quantitatively assessed using intravascular OCT-NIRF imaging. This novel theranostic approach provides a promising theranostic strategy for high-risk coronary plaques.
Collapse
Affiliation(s)
- Joon Woo Song
- Multimodal Imaging and Theranostic Lab., Cardiovascular Center, Korea University Guro Hospital, Seoul, South Korea
| | - Hyeong Soo Nam
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Jae Won Ahn
- Department of Systems Biotechnology, Chung-Ang University, Anseong, South Korea
| | - Hyun-Sang Park
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Dong Oh Kang
- Multimodal Imaging and Theranostic Lab., Cardiovascular Center, Korea University Guro Hospital, Seoul, South Korea
| | - Hyun Jung Kim
- Multimodal Imaging and Theranostic Lab., Cardiovascular Center, Korea University Guro Hospital, Seoul, South Korea
| | - Yeon Hoon Kim
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Jeongmoo Han
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Jah Yeon Choi
- Multimodal Imaging and Theranostic Lab., Cardiovascular Center, Korea University Guro Hospital, Seoul, South Korea
| | - Seung-Yul Lee
- Multimodal Imaging and Theranostic Lab., Cardiovascular Center, Korea University Guro Hospital, Seoul, South Korea
| | - Sunwon Kim
- Multimodal Imaging and Theranostic Lab., Cardiovascular Center, Korea University Guro Hospital, Seoul, South Korea
| | - Wang-Yuhl Oh
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Hongki Yoo
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Kyeongsoon Park
- Department of Systems Biotechnology, Chung-Ang University, Anseong, South Korea
| | - Jin Won Kim
- Multimodal Imaging and Theranostic Lab., Cardiovascular Center, Korea University Guro Hospital, Seoul, South Korea
| |
Collapse
|
25
|
Borchert T, Beitar L, Langer LBN, Polyak A, Wester HJ, Ross TL, Hilfiker-Kleiner D, Bengel FM, Thackeray JT. Dissecting the target leukocyte subpopulations of clinically relevant inflammation radiopharmaceuticals. J Nucl Cardiol 2021; 28:1636-1645. [PMID: 31659697 DOI: 10.1007/s12350-019-01929-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/29/2019] [Accepted: 10/01/2019] [Indexed: 10/25/2022]
Abstract
BACKGROUND Leukocyte subtypes bear distinct pro-inflammatory, reparative, and regulatory functions. Imaging inflammation provides information on disease prognosis and may guide therapy, but the cellular basis of the signal remains equivocal. We evaluated leukocyte subtype specificity of characterized clinically relevant inflammation-targeted radiotracers. METHODS AND RESULTS Leukocyte populations were purified from blood- and THP-1-derived macrophages were polarized into M1-, reparative M2a-, or M2c-macrophages. In vitro uptake assays were conducted using tracers of enhanced glucose or amino acid metabolism and molecular markers of inflammatory cells. Both 18F-deoxyglucose (18F-FDG) and the labeled amino acid 11C-methionine (11C-MET) displayed higher uptake in neutrophils and monocytes compared to other leukocytes (P = 0.005), and markedly higher accumulation in pro-inflammatory M1-macrophages compared to reparative M2a-macrophages (P < 0.001). Molecular tracers 68Ga-DOTATATE targeting the somatostatin receptor type 2 and 68Ga-pentixafor targeting the chemokine receptor type 4 (CXCR4) exhibited broad uptake by leukocyte subpopulations and polarized macrophages with highest uptake in T-cells/natural killer cells and B-cells compared to neutrophils. Mitochondrial translocator protein (TSPO)-targeted 18F-flutriciclamide selectively accumulated in monocytes and pro-inflammatory M1 macrophages (P < 0.001). Uptake by myocytes and fibroblasts tended to be higher for metabolic radiotracers. CONCLUSIONS The different in vitro cellular uptake profiles may allow isolation of distinct phases of the inflammatory pathway with specific inflammation-targeted radiotracers. The pathogenetic cell population in specific inflammatory diseases should be considered in the selection of an appropriate imaging agent.
Collapse
Affiliation(s)
- Tobias Borchert
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, 30625, Hannover, Germany
| | - Laura Beitar
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, 30625, Hannover, Germany
| | - Laura B N Langer
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, 30625, Hannover, Germany
| | - Andras Polyak
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, 30625, Hannover, Germany
| | - Hans-Jürgen Wester
- Department of Radiopharmaceutical Chemistry, Technical University of Munich, Munich, Germany
| | - Tobias L Ross
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, 30625, Hannover, Germany
| | | | - Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, 30625, Hannover, Germany
| | - James T Thackeray
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
26
|
Wang C, Leach BI, Lister D, Adams SR, Xu H, Hoh C, McConville P, Zhang J, Messer K, Ahrens ET. Metallofluorocarbon Nanoemulsion for Inflammatory Macrophage Detection via PET and MRI. J Nucl Med 2021; 62:1146-1153. [PMID: 33277399 PMCID: PMC8833871 DOI: 10.2967/jnumed.120.255273] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/12/2020] [Indexed: 12/14/2022] Open
Abstract
Inflammation is associated with a range of serious human conditions, including autoimmune and cardiovascular diseases and cancer. The ability to image active inflammatory processes greatly enhances our ability to diagnose and treat these diseases at an early stage. We describe molecular compositions enabling sensitive and precise imaging of inflammatory hotspots in vivo. Methods: A functionalized nanoemulsion with a fluorocarbon-encapsulated radiometal chelate (FERM) was developed to serve as a platform for multimodal imaging probe development. The 19F-containing FERM nanoemulsion encapsulates 89Zr in the fluorous oil via a fluorinated hydroxamic acid chelate. Simple mixing of the radiometal with the preformed aqueous nanoemulsion before use yields FERM, a stable in vivo cell tracer, enabling whole-body 89Zr PET and 19F MRI after a single intravenous injection. Results: The FERM nanoemulsion was intrinsically taken up by phagocytic immune cells, particularly macrophages, with high specificity. FERM stability was demonstrated by a high correlation between the 19F and 89Zr content in the blood (correlation coefficient > 0.99). Image sensitivity at a low dose (37 kBq) was observed in a rodent model of acute infection. The versatility of FERM was further demonstrated in models of inflammatory bowel disease and 4T1 tumor. Conclusion: Multimodal detection using FERM yields robust whole-body lesion detection and leverages the strengths of combined PET and 19F MRI. The FERM nanoemulsion has scalable production and is potentially useful for precise diagnosis, stratification, and treatment monitoring of inflammatory diseases.
Collapse
Affiliation(s)
- Chao Wang
- Department of Radiology, University of California San Diego, La Jolla, California
| | - Benjamin I Leach
- Department of Radiology, University of California San Diego, La Jolla, California
| | - Deanne Lister
- Department of Radiology, University of California San Diego, La Jolla, California
| | - Stephen R Adams
- Department of Pharmacology, University of California San Diego, La Jolla, California
| | - Hongyan Xu
- Department of Radiology, University of California San Diego, La Jolla, California
| | - Carl Hoh
- Department of Radiology, University of California San Diego, La Jolla, California
| | - Patrick McConville
- Department of Radiology, University of California San Diego, La Jolla, California
- Invicro, Inc., Boston, Massachusetts; and
| | - Jing Zhang
- Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Karen Messer
- Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Eric T Ahrens
- Department of Radiology, University of California San Diego, La Jolla, California;
| |
Collapse
|
27
|
Discovery of potential imaging and therapeutic targets for severe inflammation in COVID-19 patients. Sci Rep 2021; 11:14151. [PMID: 34239034 PMCID: PMC8266867 DOI: 10.1038/s41598-021-93743-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 06/30/2021] [Indexed: 12/15/2022] Open
Abstract
The Coronavirus disease 2019 (COVID-19) has been spreading worldwide with rapidly increased number of deaths. Hyperinflammation mediated by dysregulated monocyte/macrophage function is considered to be the key factor that triggers severe illness in COVID-19. However, no specific targeting molecule has been identified for detecting or treating hyperinflammation related to dysregulated macrophages in severe COVID-19. In this study, previously published single-cell RNA-sequencing data of bronchoalveolar lavage fluid cells from thirteen COVID-19 patients were analyzed with publicly available databases for surface and imageable targets. Immune cell composition according to the severity was estimated with the clustering of gene expression data. Expression levels of imaging target molecules for inflammation were evaluated in macrophage clusters from single-cell RNA-sequencing data. In addition, candidate targetable molecules enriched in severe COVID-19 associated with hyperinflammation were filtered. We found that expression of SLC2A3, which can be imaged by [18F]fluorodeoxyglucose, was higher in macrophages from severe COVID-19 patients. Furthermore, by integrating the surface target and drug-target binding databases with RNA-sequencing data of severe COVID-19, we identified candidate surface and druggable targets including CCR1 and FPR1 for drug delivery as well as molecular imaging. Our results provide a resource in the development of specific imaging and therapy for COVID-19-related hyperinflammation.
Collapse
|
28
|
Willemink MJ, Varga-Szemes A, Schoepf UJ, Codari M, Nieman K, Fleischmann D, Mastrodicasa D. Emerging methods for the characterization of ischemic heart disease: ultrafast Doppler angiography, micro-CT, photon-counting CT, novel MRI and PET techniques, and artificial intelligence. Eur Radiol Exp 2021; 5:12. [PMID: 33763754 PMCID: PMC7991013 DOI: 10.1186/s41747-021-00207-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 01/22/2021] [Indexed: 12/24/2022] Open
Abstract
After an ischemic event, disruptive changes in the healthy myocardium may gradually develop and may ultimately turn into fibrotic scar. While these structural changes have been described by conventional imaging modalities mostly on a macroscopic scale-i.e., late gadolinium enhancement at magnetic resonance imaging (MRI)-in recent years, novel imaging methods have shown the potential to unveil an even more detailed picture of the postischemic myocardial phenomena. These new methods may bring advances in the understanding of ischemic heart disease with potential major changes in the current clinical practice. In this review article, we provide an overview of the emerging methods for the non-invasive characterization of ischemic heart disease, including coronary ultrafast Doppler angiography, photon-counting computed tomography (CT), micro-CT (for preclinical studies), low-field and ultrahigh-field MRI, and 11C-methionine positron emission tomography. In addition, we discuss new opportunities brought by artificial intelligence, while addressing promising future scenarios and the challenges for the application of artificial intelligence in the field of cardiac imaging.
Collapse
Affiliation(s)
- Martin J. Willemink
- grid.168010.e0000000419368956Department of Radiology, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94035 USA
| | - Akos Varga-Szemes
- grid.259828.c0000 0001 2189 3475Division of Cardiovascular Imaging, Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, SC USA
| | - U. Joseph Schoepf
- grid.259828.c0000 0001 2189 3475Division of Cardiovascular Imaging, Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, SC USA
| | - Marina Codari
- grid.168010.e0000000419368956Department of Radiology, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94035 USA
| | - Koen Nieman
- grid.168010.e0000000419368956Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA ,Stanford Cardiovascular Institute, Stanford, CA 94305 USA
| | - Dominik Fleischmann
- grid.168010.e0000000419368956Department of Radiology, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94035 USA ,Stanford Cardiovascular Institute, Stanford, CA 94305 USA
| | - Domenico Mastrodicasa
- Department of Radiology, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94035, USA. .,Stanford Cardiovascular Institute, Stanford, CA, 94305, USA.
| |
Collapse
|
29
|
Imaging Inflammation with Positron Emission Tomography. Biomedicines 2021; 9:biomedicines9020212. [PMID: 33669804 PMCID: PMC7922638 DOI: 10.3390/biomedicines9020212] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 01/28/2021] [Accepted: 02/12/2021] [Indexed: 12/19/2022] Open
Abstract
The impact of inflammation on the outcome of many medical conditions such as cardiovascular diseases, neurological disorders, infections, cancer, and autoimmune diseases has been widely acknowledged. However, in contrast to neurological, oncologic, and cardiovascular disorders, imaging plays a minor role in research and management of inflammation. Imaging can provide insights into individual and temporospatial biology and grade of inflammation which can be of diagnostic, therapeutic, and prognostic value. There is therefore an urgent need to evaluate and understand current approaches and potential applications for imaging of inflammation. This review discusses radiotracers for positron emission tomography (PET) that have been used to image inflammation in cardiovascular diseases and other inflammatory conditions with a special emphasis on radiotracers that have already been successfully applied in clinical settings.
Collapse
|
30
|
Chaudhry F, Kawai H, Johnson KW, Narula N, Shekhar A, Chaudhry F, Nakahara T, Tanimoto T, Kim D, Adapoe MKMY, Blankenberg FG, Mattis JA, Pak KY, Levy PD, Ozaki Y, Arbustini E, Strauss HW, Petrov A, Fuster V, Narula J. Molecular Imaging of Apoptosis in Atherosclerosis by Targeting Cell Membrane Phospholipid Asymmetry. J Am Coll Cardiol 2021; 76:1862-1874. [PMID: 33059832 DOI: 10.1016/j.jacc.2020.08.047] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 08/20/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Apoptosis in atherosclerotic lesions contributes to plaque vulnerability by lipid core enlargement and fibrous cap attenuation. Apoptosis is associated with exteriorization of phosphatidylserine (PS) and phosphatidylethanolamine (PE) on the cell membrane. Although PS-avid radiolabeled annexin-V has been employed for molecular imaging of high-risk plaques, PE-targeted imaging in atherosclerosis has not been studied. OBJECTIVES This study sought to evaluate the feasibility of molecular imaging with PE-avid radiolabeled duramycin in experimental atherosclerotic lesions in a rabbit model and compare duramycin targeting with radiolabeled annexin-V. METHODS Of the 27 rabbits, 21 were fed high-cholesterol, high-fat diet for 16 weeks. Nine of the 21 rabbits received 99mTc-duramycin (test group), 6 received 99mTc-linear duramycin (duramycin without PE-binding capability, negative radiotracer control group), and 6 received 99mTc-annexin-V for radionuclide imaging. The remaining normal chow-fed 6 animals (disease control group) received 99mTc-duramycin. In vivo microSPECT/microCT imaging was performed, and the aortas were explanted for ex vivo imaging and for histological characterization of atherosclerosis. RESULTS A significantly higher duramycin uptake was observed in the test group compared with that of disease control and negative radiotracer control animals; duramycin uptake was also significantly higher than the annexin-V uptake. Quantitative duramycin uptake, represented as the square root of percent injected dose per cm (√ID/cm) of abdominal aorta was >2-fold higher in atherosclerotic lesions in test group (0.08 ± 0.01%) than in comparable regions of disease control animals (0.039 ± 0.0061%, p = 3.70·10-8). Mean annexin uptake (0.060 ± 0.010%) was significantly lower than duramycin (p = 0.001). Duramycin uptake corresponded to the lesion severity and macrophage burden. The radiation burden to the kidneys was substantially lower with duramycin (0.49% ID/g) than annexin (5.48% ID/g; p = 4.00·10-4). CONCLUSIONS Radiolabeled duramycin localizes in lipid-rich areas with high concentration of apoptotic macrophages in the experimental atherosclerosis model. Duramycin uptake in atherosclerotic lesions was significantly greater than annexin-V uptake and produced significantly lower radiation burden to nontarget organs.
Collapse
Affiliation(s)
- Farhan Chaudhry
- Icahn School of Medicine at Mount Sinai, New York, New York; Wayne State University School of Medicine, Detroit, Michigan
| | - Hideki Kawai
- Icahn School of Medicine at Mount Sinai, New York, New York; Department of Cardiology, Fujita Health University, Toyoake, Aichi, Japan
| | - Kipp W Johnson
- Icahn School of Medicine at Mount Sinai, New York, New York
| | - Navneet Narula
- New York University Langone Medical Center, New York, New York
| | - Aditya Shekhar
- Icahn School of Medicine at Mount Sinai, New York, New York
| | | | | | | | - Dongbin Kim
- Icahn School of Medicine at Mount Sinai, New York, New York
| | | | | | - Jeffrey A Mattis
- Molecular Targeting Technologies, Inc., West Chester, Pennsylvania
| | - Koon Y Pak
- Molecular Targeting Technologies, Inc., West Chester, Pennsylvania
| | - Phillip D Levy
- Wayne State University School of Medicine, Detroit, Michigan
| | - Yukio Ozaki
- Department of Cardiology, Fujita Health University, Toyoake, Aichi, Japan
| | | | - H William Strauss
- Icahn School of Medicine at Mount Sinai, New York, New York; Memorial Sloan Kettering Cancer Center, New York, New York
| | - Artiom Petrov
- Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Valentin Fuster
- Icahn School of Medicine at Mount Sinai, New York, New York; Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Jagat Narula
- Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
31
|
Varasteh Z, De Rose F, Mohanta S, Li Y, Zhang X, Miritsch B, Scafetta G, Yin C, Sager HB, Glasl S, Gorpas D, Habenicht AJ, Ntziachristos V, Weber WA, Bartolazzi A, Schwaiger M, D'Alessandria C. Imaging atherosclerotic plaques by targeting Galectin-3 and activated macrophages using ( 89Zr)-DFO- Galectin3-F(ab') 2 mAb. Am J Cancer Res 2021; 11:1864-1876. [PMID: 33408786 PMCID: PMC7778602 DOI: 10.7150/thno.50247] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 11/11/2020] [Indexed: 12/12/2022] Open
Abstract
Rationale: The high expression of Galectin-3 (Gal3) in macrophages of atherosclerotic plaques suggests its participation in atherosclerosis pathogenesis, and raises the possibility to use it as a target to image disease severity in vivo. Here, we explored the feasibility of tracking atherosclerosis by targeting Gal3 expression in plaques of apolipoprotein E knockout (ApoE-KO) mice via PET imaging. Methods: Targeting of Gal3 in M0-, M1- and M2 (M2a/M2c)-polarized macrophages was assessed in vitro using a Gal3-F(ab')2 mAb labeled with AlexaFluor®488 and 89Zr- desferrioxamine-thioureyl-phenyl-isothiocyanate (DFO). To visualize plaques in vivo, ApoE-KO mice were injected i.v. with 89Zr-DFO-Gal3-F(ab')2 mAb and imaged via PET/CT 48 h post injection. Whole length aortas harvested from euthanized mice were processed for Sudan-IV staining, autoradiography, and immunostaining for Gal3, CD68 and α-SMA expression. To confirm accumulation of the tracer in plaques, ApoE-KO mice were injected i.v. with Cy5.5-Gal3-F(ab')2 mAb, euthanized 48 h post injection, followed by cryosections of the body and acquisition of fluorescent images. To explore the clinical potential of this imaging modality, immunostaining for Gal3, CD68 and α-SMA expression were carried out in human plaques. Single cell RNA sequencing (scRNA-Seq) analyses were performed to measure LGALS3 (i.e. a synonym for Gal3) gene expression in each macrophage of several subtypes present in murine or human plaques. Results: Preferential binding to M2 macrophages was observed with both AlexaFluor®488-Gal3-F(ab')2 and 89Zr-DFO-Gal3-F(ab')2 mAbs. Focal and specific 89Zr-DFO-Gal3-F(ab')2 mAb uptake was detected in plaques of ApoE-KO mice by PET/CT. Autoradiography and immunohistochemical analyses of aortas confirmed the expression of Gal3 within plaques mainly in macrophages. Moreover, a specific fluorescent signal was visualized within the lesions of vascular structures burdened by plaques in mice. Gal3 expression in human plaques showed similar Gal3 expression patterns when compared to their murine counterparts. Conclusions: Our data reveal that 89Zr-DFO-Gal3-F(ab')2 mAb PET/CT is a potentially novel tool to image atherosclerotic plaques at different stages of development, allowing knowledge-based tailored individual intervention in clinically significant disease.
Collapse
|
32
|
Osborn EA, Albaghdadi M, Libby P, Jaffer FA. Molecular Imaging of Atherosclerosis. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00086-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
33
|
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: 3.3] [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
|
34
|
Tahara N, Nitta Y, Bekki M, Tahara A, Maeda-Ogata S, Sugiyama Y, Honda A, Igata S, Nakamura T, Sun J, Kurata S, Fujimoto K, Abe T, Matsui T, Yamagishi SI, Fukumoto Y. Two-hour postload plasma glucose and pigment epithelium-derived factor levels are markers of coronary artery inflammation in type 2 diabetic patients. J Nucl Cardiol 2020; 27:1352-1364. [PMID: 31407236 DOI: 10.1007/s12350-019-01842-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 07/08/2019] [Indexed: 11/28/2022]
Abstract
BACKGROUND We have previously found that pioglitazone attenuates inflammation in the left main trunk of coronary artery (LMT), evaluated as target-to-background ratio (TBR) by 18F-fluorodeoxyglucose-positron emission tomography/computed tomography (FDG-PET/CT) in patients with impaired glucose tolerance or type 2 diabetes. OBJECTIVES We assessed which clinical variables could predict the change in TBR in the LMT after 4-month add-on therapy with oral hypoglycemic agents (OHAs). METHODS A total of 38 type 2 diabetic patients with carotid atherosclerosis who had already received OHAs except for pioglitazone was enrolled. At baseline and 4 months after add-on therapy with pioglitazone or glimepiride, all patients underwent 75 g oral glucose tolerance test, blood chemistry analysis, and FDG-PET/CT. RESULTS Fasting plasma glucose, 30-, 60-, 90-, 120-minutes postload plasma glucose, HbA1c, and LMT-TBR values were significantly decreased by add-on therapy, whereas high-density lipoprotein-cholesterol and adiponectin levels were increased. Increased serum levels of pigment epithelium-derived factor (PEDF), a marker of insulin resistance and non-use of aspirin at baseline could predict the favorable response of LMT-TBR to add-on therapy. Moreover, Δ120-minutes postload plasma glucose and ΔPEDF were independent correlates of ΔLMT-TBR. CONCLUSIONS Our present study suggests that 120-minutes postload plasma glucose and PEDF values may be markers and potential therapeutic targets of coronary artery inflammation in type 2 diabetic patients. CLINICAL TRIAL REGISTRATION URL: http://clinicaltrials.gov . Unique identifier: NCT00722631. New markers for diabetes and CAD is on the horizon! Two-hour postload plasma glucose and pigment epithelium derived factor are markers of coronary artery inflammation in type 2 diabetic patients.
Collapse
Affiliation(s)
- Nobuhiro Tahara
- Division of Cardiovascular Medicine, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan.
| | - Yoshikazu Nitta
- Division of Cardiovascular Medicine, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan
| | - Munehisa Bekki
- Division of Cardiovascular Medicine, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan
| | - Atsuko Tahara
- Division of Cardiovascular Medicine, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan
| | - Shoko Maeda-Ogata
- Division of Cardiovascular Medicine, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan
| | - Yoichi Sugiyama
- Division of Cardiovascular Medicine, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan
| | - Akihiro Honda
- Division of Cardiovascular Medicine, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan
| | - Sachiyo Igata
- Division of Cardiovascular Medicine, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan
| | - Tomohisa Nakamura
- Division of Cardiovascular Medicine, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan
| | - Jiahui Sun
- Division of Cardiovascular Medicine, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan
| | - Seiji Kurata
- Department of Radiology, Kurume University School of Medicine, Kurume, Japan
| | - Kiminori Fujimoto
- Department of Radiology, Kurume University School of Medicine, Kurume, Japan
| | - Toshi Abe
- Department of Radiology, Kurume University School of Medicine, Kurume, Japan
| | - Takanori Matsui
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume, Japan
| | - Sho-Ichi Yamagishi
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Tokyo, 142-8666, Japan
| | - Yoshihiro Fukumoto
- Division of Cardiovascular Medicine, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan
| |
Collapse
|
35
|
Vigne J, Hyafil F. Inflammation imaging to define vulnerable plaque or vulnerable patient. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2020; 64:21-34. [DOI: 10.23736/s1824-4785.20.03231-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
36
|
Abstract
Most of the acute ischemic events, such as acute coronary syndromes and stroke, are attributed to vulnerable plaques. These lesions have common histological and pathophysiological features, including inflammatory cell infiltration, neo-angiogenesis, remodelling, haemorrhage predisposition, thin fibrous cap, large lipid core, and micro-calcifications. Early detection of the presence of a plaque prone to rupture could be life-saving for the patient; however, vulnerable plaques usually cause non-haemodynamically significant stenosis, and anatomical imaging techniques often underestimate, or may not even detect, these lesions. Although ultrasound techniques are currently considered as the "first-line" examinations for the diagnostic investigation and treatment monitoring in patients with atherosclerotic plaques, positron emission tomography (PET) imaging could open new horizons in the assessment of atherosclerosis, given its ability to visualize metabolic processes and provide molecular-functional evidence regarding vulnerable plaques. Moreover, modern hybrid imaging techniques, combining PET with computed tomography or magnetic resonance imaging, can evaluate simultaneously both functional and morphological parameters of the atherosclerotic plaques, and are expected to significantly expand their clinical role in the future. This review summarizes current research on the PET imaging of the vulnerable atherosclerotic plaques, outlining current and potential applications in the clinical setting.
Collapse
|
37
|
Abstract
Atherosclerosis is a chronic and most often progressive disease with a long clinically apparently silent period, and can become unstable at any time, due to a plaque rupture or erosion, leading to an acute atherothrombotic event. Atherosclerosis has a progression rate that is highly variable among patients and in the same patient. The progression of atherosclerotic plaque from asymptomatic to symptomatic phase depends on its structure and composition in which inflammation plays an essential role. Prototype of the ruptured plaque contains a large, soft, lipid-rich necrotic core with intraplaque hemorrhage that accounts for more than half of the volume of the plaque covered by a thin and inflamed fibrous cap with few smooth muscle cells, and a heavy infiltrate of inflammatory cells. Noninvasive imaging modalities might provide an assessment of the atherosclerotic disease process through the exploration of these plaque features. Computed tomography angiography and magnetic resonance imaging can characterize plaque morphology, whereas molecular imaging, owing to the high sensitivity of nuclear medicine for the detection of radiopharmaceuticals in tissues, allows to explore plaque biology. During the last 2 decades, FDG-PET imaging has also emerged as a powerful tool to explore noninvasively inflammatory activities in atherosclerotic plaques providing new insights on the evolution of metabolic activities in the vascular wall over time. This review highlights the role of PET imaging for the exploration of metabolic activities in atherosclerotic plaques. It will resume the evidence that have been gathered from clinical studies using FDG-PET and will discuss the perspectives of new radiopharmaceuticals for vulnerable plaque imaging.
Collapse
Affiliation(s)
- Olivier Lairez
- Cardiac Imaging Centre, Rangueil University Hospital, Toulouse, France
| | - Fabien Hyafil
- Department of Nuclear Medicine, Bichat University Hospital, Hôpitaux de Paris, Université René Diderot, Paris, France.
| |
Collapse
|
38
|
Heo GS, Sultan D, Liu Y. Current and novel radiopharmaceuticals for imaging cardiovascular inflammation. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2020; 64:4-20. [PMID: 32077667 DOI: 10.23736/s1824-4785.20.03230-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Cardiovascular disease (CVD) remains the leading cause of death worldwide despite advances in diagnostic technologies and treatment strategies. The underlying cause of most CVD is atherosclerosis, a chronic disease driven by inflammatory reactions. Atherosclerotic plaque rupture could cause arterial occlusion leading to ischemic tissue injuries such as myocardial infarction (MI) and stroke. Clinically, most imaging modalities are based on anatomy and provide limited information about the on-going molecular activities affecting the vulnerability of atherosclerotic lesion for risk stratification of patients. Thus, the ability to differentiate stable plaques from those that are vulnerable is an unmet clinical need. Of various imaging techniques, the radionuclide-based molecular imaging modalities including positron emission tomography and single-photon emission computerized tomography provide superior ability to noninvasively visualize molecular activities in vivo and may serve as a useful tool in tackling this challenge. Moreover, the well-established translational pathway of radiopharmaceuticals may also facilitate the translation of discoveries from benchtop to clinical investigation in contrast to other imaging modalities to fulfill the goal of precision medicine. The relationship between inflammation occurring within the plaque and its proneness to rupture has been well documented. Therefore, an active effort has been significantly devoted to develop radiopharmaceuticals specifically to measure CVD inflammatory status, and potentially elucidate those plaques which are prone to rupture. In the following review, molecular imaging of inflammatory biomarkers will be briefly discussed.
Collapse
Affiliation(s)
- Gyu S Heo
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO, USA
| | - Deborah Sultan
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO, USA
| | - Yongjian Liu
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO, USA -
| |
Collapse
|
39
|
Iking J, Klose J, Staniszewska M, Fendler WP, Herrmann K, Rischpler C. Imaging inflammation after myocardial infarction: implications for prognosis and therapeutic guidance. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2020; 64:35-50. [PMID: 32077669 DOI: 10.23736/s1824-4785.20.03232-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Inflammation after myocardial infarction (MI) has been in the focus of cardiovascular research for several years as it influences the remodeling process of the ischemic heart and thereby critically determines the clinical outcome of the patient. Today, it is well appreciated that inflammation is a crucial necessity for the initiation of the natural wound healing process; however, excessive inflammation can have detrimental effects and might result in adverse ventricular remodeling which is associated with an increased risk of heart failure. Newly emerged imaging techniques facilitate the non-invasive assessment of immune cell infiltration into the ischemic myocardium and can provide greater insight into the underlying complex and dynamic repair mechanisms. Molecular imaging of inflammation in the context of MI may help with stratification of patients at high risk of adverse ventricular remodeling post-MI which may be of diagnostic, therapeutic, and prognostic value. Novel radiopharmaceuticals may additionally provide a way to combine patient monitoring and therapy. In spite of great advances in recent years in the field of imaging sciences, clinicians still need to overcome some obstacles to a wider implementation of inflammation imaging post-MI. This review focuses on inflammation as a molecular imaging target and its potential implication in prognosis and therapeutic guidance.
Collapse
Affiliation(s)
- Janette Iking
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany.,Department of Cardiology I for Coronary and Peripheral Vascular Disease, and Heart Failure, University Hospital Münster, Münster, Germany
| | - Jasmin Klose
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | | | - Wolfgang P Fendler
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | | |
Collapse
|
40
|
Abstract
This review discusses nuclear imaging of inflammation using molecular probes beyond fluoro-d-glucose, is structured by cellular targets, and focuses on those tracers that have been successfully applied clinically.
Collapse
Affiliation(s)
- Malte Kircher
- Department of Nuclear Medicine, University Hospital Augsburg, Stenglinstr. 2, Würzburg 86156, Germany
| | - Constantin Lapa
- Department of Nuclear Medicine, University Hospital Augsburg, Stenglinstr. 2, Würzburg 86156, Germany.
| |
Collapse
|
41
|
Pérez-Medina C, Fayad ZA, Mulder WJM. Atherosclerosis Immunoimaging by Positron Emission Tomography. Arterioscler Thromb Vasc Biol 2020; 40:865-873. [PMID: 32078338 DOI: 10.1161/atvbaha.119.313455] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The immune system's role in atherosclerosis has long been an important research topic and is increasingly investigated for therapeutic and diagnostic purposes. Therefore, noninvasive imaging of hematopoietic organs and immune cells will undoubtedly improve atherosclerosis phenotyping and serve as a monitoring method for immunotherapeutic treatments. Among the available imaging techniques, positron emission tomography's unique features make it an ideal tool to quantitatively image the immune response in the context of atherosclerosis and afford reliable readouts to guide medical interventions in cardiovascular disease. Here, we summarize the state of the art in the field of atherosclerosis positron emission tomography immunoimaging and provide an outlook on current and future applications.
Collapse
Affiliation(s)
- Carlos Pérez-Medina
- From the Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (C.P.-M.).,Icahn School of Medicine at Mount Sinai, New York (C.P.-M., Z.A.F., W.J.M.M.)
| | - Zahi A Fayad
- Icahn School of Medicine at Mount Sinai, New York (C.P.-M., Z.A.F., W.J.M.M.)
| | - Willem J M Mulder
- Icahn School of Medicine at Mount Sinai, New York (C.P.-M., Z.A.F., W.J.M.M.).,Eindhoven University of Technology, the Netherlands (W.J.M.M.)
| |
Collapse
|
42
|
Thackeray JT, Bengel FM. Molecular Imaging of Myocardial Inflammation With Positron Emission Tomography Post-Ischemia: A Determinant of Subsequent Remodeling or Recovery. JACC Cardiovasc Imaging 2019; 11:1340-1355. [PMID: 30190033 DOI: 10.1016/j.jcmg.2018.05.026] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 05/09/2018] [Accepted: 05/12/2018] [Indexed: 12/20/2022]
Abstract
Inflammation after myocardial ischemia influences ventricular remodeling and repair and has emerged as a therapeutic target. Conventional diagnostic measurements address systemic inflammation but cannot quantify local tissue changes. Molecular imaging facilitates noninvasive assessment of leukocyte infiltration into damaged myocardium. Preliminary experience with 18F-labeled fluorodeoxyglucose ([18F]FDG) demonstrates localized inflammatory cell signal within the infarct territory as an independent predictor of subsequent ventricular dysfunction. Novel targeted radiotracers may provide additional insight into the enrichment of specific leukocyte populations. Challenges to wider implementation of inflammation imaging after myocardial infarction include accurate and reproducible quantification, prognostic value, and capacity to monitor inflammation response to novel treatment. This review describes myocardial inflammation following ischemia as a molecular imaging target and evaluates established and emerging radiotracers for this application. Furthermore, the potential role of inflammation imaging to provide prognostic information, support novel drug and therapeutic research, and assess biological response to cardiac disease is discussed.
Collapse
Affiliation(s)
- James T Thackeray
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany.
| | - Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| |
Collapse
|
43
|
Ohashi R, Fukazawa R, Shimizu A, Ogawa S, Ochi M, Nitta T, Itoh Y. M1 macrophage is the predominant phenotype in coronary artery lesions following Kawasaki disease. Vasc Med 2019; 24:484-492. [PMID: 31621532 DOI: 10.1177/1358863x19878495] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Kawasaki disease (KD) is a systemic inflammatory process that affects the medium-sized arteries, causing various cardiovascular complications. However, it is not clear if the vascular sequelae following KD can predispose to the development of atherosclerosis later in life. Our aim was to examine the macrophage phenotypes in the coronary arteries forming giant aneurysms after KD to gain insight into the pathogenesis of vascular lesions in KD. We examined histological sections of the coronary arteries from five patients with KD who underwent coronary bypass grafting procedure as treatment for giant aneurysms and subsequent stenosis. Immunohistochemical expression of M1- and M2-macrophage markers was assessed to determine the macrophage phenotype of KD to compare with that of atherosclerosis in eight adult patients. All the KD specimens showed a mild to moderate degree of intimal thickening consisting of mature fibrous tissue and distortion of elastic fibers, mimicking the histological features of atherosclerosis. The total number of CD68 positive macrophages was higher in atherosclerosis than in KD specimens. Among the CD68 positive macrophages, the proportion of M1 phenotype, detected by CD86 or SOCS3, was higher in KD than in atherosclerosis. In contrast, the proportion of M2 phenotype, detected by CD163 or MRC1, was higher in patients with atherosclerosis. Despite similar histological features, KD and atherosclerosis appear to have a different immunological etiology for progression of the chronic vascular lesions. A further study enrolling a larger number of cases is required to delineate underlying mechanisms of vascular complications in KD.
Collapse
Affiliation(s)
- Ryuji Ohashi
- Department of Diagnostic Pathology, Nippon Medical School Musashi-kosugi Hospital, Nakahara-ku, Kanagawa, Japan
| | - Ryuji Fukazawa
- Department of Pediatrics, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| | - Akira Shimizu
- Department of Analytic Human Pathology, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| | - Shunichi Ogawa
- Department of Pediatrics, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| | - Masami Ochi
- Department of Cardiovascular Surgery, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| | - Takashi Nitta
- Department of Cardiovascular Surgery, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| | - Yasuhiko Itoh
- Department of Pediatrics, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
44
|
Mukherjee S, Sonanini D, Maurer A, Daldrup-Link HE. The yin and yang of imaging tumor associated macrophages with PET and MRI. Am J Cancer Res 2019; 9:7730-7748. [PMID: 31695797 PMCID: PMC6831464 DOI: 10.7150/thno.37306] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 08/27/2019] [Indexed: 12/14/2022] Open
Abstract
Tumor associated macrophages (TAM) are key players in the cancer microenvironment. Molecular imaging modalities such as MRI and PET can be used to track and monitor TAM dynamics in tumors non-invasively, based on specific uptake and quantification of MRI-detectable nanoparticles or PET-detectable radiotracers. Particular molecular signatures can be leveraged to target anti-inflammatory TAM, which support tumor growth, and pro-inflammatory TAM, which suppress tumor growth. In addition, TAM-directed imaging probes can be designed to include immune modulating properties, thereby leading to combined diagnostic and therapeutic (theranostic) effects. In this review, we will discuss the complementary role of TAM-directed radiotracers and iron oxide nanoparticles for monitoring cancer immunotherapies with PET and MRI technologies. In addition, we will outline how TAM-directed imaging and therapy is interdependent and can be connected towards improved clinical outcomes
Collapse
|
45
|
Narula J, Arbustini E. Inflammation, Superadded Inflammation, and Out-of-Proportion Inflammation in Atherosclerosis. JAMA Cardiol 2019; 3:912-914. [PMID: 30208401 DOI: 10.1001/jamacardio.2018.2760] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Jagat Narula
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Eloisa Arbustini
- Center for Inherited Cardiovascular Diseases, Istituto di Ricovero e Cura a Carattere Scientifico Fondazione Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
46
|
Fayad ZA, Swirski FK, Calcagno C, Robbins CS, Mulder W, Kovacic JC. Monocyte and Macrophage Dynamics in the Cardiovascular System: JACC Macrophage in CVD Series (Part 3). J Am Coll Cardiol 2019; 72:2198-2212. [PMID: 30360828 DOI: 10.1016/j.jacc.2018.08.2150] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 07/16/2018] [Accepted: 08/03/2018] [Indexed: 12/12/2022]
Abstract
It has long been recognized that the bone marrow is the primary site of origin for circulating monocytes that may later become macrophages in atherosclerotic lesions. However, only in recent times has the complex relationship among the bone marrow, monocytes/macrophages, and atherosclerotic plaques begun to be understood. Moreover, the systemic nature of these interactions, which also involves additional compartments such as extramedullary hematopoietic sites (i.e., spleen), is only just becoming apparent. In parallel, progressive advances in imaging and cell labeling techniques have opened new opportunities for in vivo imaging of monocyte/macrophage trafficking in atherosclerotic lesions and at the systemic level. In this Part 3 of a 4-part review series covering the macrophage in cardiovascular disease, the authors intersect systemic biology with advanced imaging techniques to explore monocyte and macrophage dynamics in the cardiovascular system, with an emphasis on how events at the systemic level might affect local atherosclerotic plaque biology.
Collapse
Affiliation(s)
- Zahi A Fayad
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, New York; The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Filip K Swirski
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Claudia Calcagno
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Clinton S Robbins
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts; Peter Munk Cardiac Centre, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada; Departments of Laboratory Medicine and Pathobiology and Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Willem Mulder
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jason C Kovacic
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
47
|
Abstract
Noninvasive imaging technologies offer to identify several anatomic and molecular features of high-risk plaques. For the noninvasive molecular imaging of atherosclerotic plaques, nuclear medicine constitutes one of the best imaging modalities, thanks to its high sensitivity for the detection of probes in tissues. 18F-fluorodeoxyglucose (FDG) is currently the most widely used radiopharmaceutical for molecular imaging of atherosclerotic plaques with positron emission tomography. The intensity of FDG uptake in the vascular wall correlates closely with the degree of macrophage infiltration in atherosclerotic plaques. FDG positron emission tomographic imaging has become a powerful tool to identify and monitor noninvasively inflammatory activities in atherosclerotic plaques over time. This review examines how FDG positron emission tomographic imaging has given us deeper insight into the role of inflammation in atherosclerotic plaque progression and discusses perspectives for alternative radiopharmaceuticals to FDG that could provide a more specific and simple identification of high-risk lesions and help improve risk stratification of atherosclerotic patients.
Visual Overview—
An online visual overview is available for this article.
Collapse
Affiliation(s)
- Fabien Hyafil
- From the Department of Nuclear Medicine, Bichat University Hospital, Assistance Publique–Hôpitaux de Paris (F.H.), University Paris 7 René Diderot, France
- INSERM U1148, Laboratory for Vascular Translational Science, DHU FIRE (F.H., J.V.), University Paris 7 René Diderot, France
| | - Jonathan Vigne
- INSERM U1148, Laboratory for Vascular Translational Science, DHU FIRE (F.H., J.V.), University Paris 7 René Diderot, France
- Department of Nuclear Medicine, CHU de Caen Normandie, Normandie University, UNICAEN, France (J.V.)
| |
Collapse
|
48
|
Abstract
Noninvasive imaging has played an increasing role in the process of cardiovascular drug development. This review focuses specifically on the use of molecular imaging, which has been increasingly applied to improve and accelerate certain preclinical steps in drug development, including the identification of appropriate therapeutic targets, evaluation of on-target and off-target effects of candidate therapies, assessment of dose response, and the evaluation of drug or biological biodistribution and pharmacodynamics. Unlike the case in cancer medicine, in cardiovascular medicine, molecular imaging has not been used as a primary surrogate clinical end point for drug approval. However, molecular imaging has been applied in early clinical trials, particularly in phase 0 studies, to demonstrate proof-of-concept or to explain variation in treatment effect. Many of these applications where molecular imaging has been used in drug development have involved the retasking of technologies that were originally intended as clinical diagnostics. With greater experience and recognition of the rich information provided by in vivo molecular imaging, it is anticipated that it will increasingly be used to address the enormous time and costs associated with bringing a new drug to clinical launch.
Collapse
Affiliation(s)
- Jonathan R Lindner
- From the Knight Cardiovascular Institute (J.R.L.), Oregon National Primate Research Center (J.R.L.), and Center for Radiologic Research (J.L.), Oregon Health and Science University, Portland.
| | - Jeanne Link
- From the Knight Cardiovascular Institute (J.R.L.), Oregon National Primate Research Center (J.R.L.), and Center for Radiologic Research (J.L.), Oregon Health and Science University, Portland
| |
Collapse
|
49
|
Lavin Plaza B, Theodoulou I, Rashid I, Hajhosseiny R, Phinikaridou A, Botnar RM. Molecular Imaging in Ischemic Heart Disease. CURRENT CARDIOVASCULAR IMAGING REPORTS 2019; 12:31. [PMID: 31281564 PMCID: PMC6557873 DOI: 10.1007/s12410-019-9500-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Purpose of Review The purpose of this paper is to review current and new modalities to image key biological processes in ischemic heart disease and after myocardial infarction non-invasively. Recent Findings New imaging targets have been developed to detect and quantify myocardial damage after ischemia. Although positron emission tomography (PET) has been leading the development of new probes in the past, continuous improvements of magnetic resonance imaging (MRI) together with the development of new novel MRI contrast agents opens new research avenues including the combination of both PET and MRI to obtain anatomic, functional, and molecular information simultaneously, which is not possible from a single imaging session. Summary This review summarizes the state of art of non-invasive molecular imaging of the myocardium during ischemia and after myocardial infarction using PET and MRI. We also describe the different contrast agents that have been developed to image the different phases of cardiac healing and the biological processes associated with each of those phases. Importantly, here we focus on imaging of inflammation as it is the key biological process that orchestrates clearance of dead cells, tissue remodeling, cardiac repair, and future outcome. We also focus on clinical translation of some of the novel contrast agents that have been tested in patients and discuss the need for larger, multi-center patient studies to fully validate the applicability of new imaging probes.
Collapse
Affiliation(s)
- Begoña Lavin Plaza
- 1School of Biomedical Engineering and Imaging Sciences, King's College London, 3rd Floor, Lambeth wing, St Thomas Hospital, London, SE1 7EH UK
| | - Iakovos Theodoulou
- 2Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Imran Rashid
- 1School of Biomedical Engineering and Imaging Sciences, King's College London, 3rd Floor, Lambeth wing, St Thomas Hospital, London, SE1 7EH UK
| | - Reza Hajhosseiny
- 1School of Biomedical Engineering and Imaging Sciences, King's College London, 3rd Floor, Lambeth wing, St Thomas Hospital, London, SE1 7EH UK
| | - Alkystis Phinikaridou
- 1School of Biomedical Engineering and Imaging Sciences, King's College London, 3rd Floor, Lambeth wing, St Thomas Hospital, London, SE1 7EH UK
| | - Rene M Botnar
- 1School of Biomedical Engineering and Imaging Sciences, King's College London, 3rd Floor, Lambeth wing, St Thomas Hospital, London, SE1 7EH UK.,3Escuela de Ingeniería, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
50
|
Synergistic protective effects of a statin and an angiotensin receptor blocker for initiation and progression of atherosclerosis. PLoS One 2019; 14:e0215604. [PMID: 31050669 PMCID: PMC6499436 DOI: 10.1371/journal.pone.0215604] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 04/05/2019] [Indexed: 12/31/2022] Open
Abstract
Aim Although the atheroprotective effects of statins and angiotensin II receptor blockers (ARBs) are well-established, little is known about their additive effects, especially during the early period of atherosclerosis. The aim of this study was to investigate whether combination of a statin and an ARB exerts synergistic anti-atherosclerotic effects, and to elucidate the mechanisms of combined effects. Methods Atherosclerotic plaques were developed in arteries of 23 rabbits using a high-cholesterol diet (HCD) and intra-arterial balloon inflation. Rabbits received one of five different treatment strategies for 4 weeks: positive control [n = 5, HCD]; negative control [n = 3, regular chow diet]; statin [n = 5, HCD and rosuvastatin 10 mg]; ARB [n = 5, HCD and olmesartan 20 mg]; and combination [n = 5, HCD and statin+ARB]. Results Histological analysis demonstrated that development of atherosclerotic plaques was inhibited more in combination group than in statin group (P = 0.001). Although macrophage infiltration identified by RAM11 staining was not significantly different between combination and individual treatment groups (31.76±4.84% [combination] vs. 38.11±6.53% [statin; P = 0.35] or 35.14±2.87% [ARB; P = 0.62]), the relative proportion of pro-inflammatory M1-macrophages was significantly lower in combination group than in ARB group (3.20±0.47% vs. 5.20±0.78%, P = 0.02). Furthermore, M2-macrophage polarization was higher in combination group than in statin group (17.70±3.04% vs. 7.86±0.68%, P = 0.001). Conclusion Combination treatment with a statin and an ARB produced synergistic protective effects for atherosclerosis initiation and progression, which may be attributed to modulation of macrophage characteristics in the early period of atherosclerosis.
Collapse
|