1
|
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
|
2
|
Higuchi T, Serfling SE, Leistner DM, Speer T, Werner RA. FAPI-PET in Cardiovascular Disease. Semin Nucl Med 2024; 54:747-752. [PMID: 38519308 DOI: 10.1053/j.semnuclmed.2024.02.006] [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: 02/19/2024] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/24/2024]
Abstract
PET probes targeting fibroblasts are frequently used for varying applications in oncology. In recent years, the clinical spectrum has been expanded towards cardiovascular medicine, e.g., after myocardial infarction, in aortic stenosis or as a non-invasive read-out of atherosclerosis. We herein provide a brief overview of the current status of this PET radiotracer in the context of cardiovascular disease, including translational and clinical evidence. In addition, we will also briefly discuss future applications, e.g., the use of fibroblast-targeting PET to investigate bilateral organ function along the cardiorenal axis.
Collapse
Affiliation(s)
- Takahiro Higuchi
- Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg; Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | | | - David M Leistner
- Department of Cardiology/Angiology, University Heart Center Frankfurt, Goethe University Hospital, Frankfurt, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Frankfurt Rhine-Main, Frankfurt, Germany
| | - Thimoteus Speer
- Department of Internal Medicine 4 - Nephrology, Goethe University Frankfurt, Frankfurt am Main, Germany; Else Kröner-Fresenius-Zentrum for Nephrological Research, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Rudolf A Werner
- Goethe University Frankfurt, University Hospital, Department of Nuclear Medicine, Clinic for Radiology and Nuclear Medicine, Germany; Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD.
| |
Collapse
|
3
|
Li J, Hong X, Chen Y, Yin B, Yang H, Shi C, Zeng X, Zhang D, Guo Z, Zhang X. MicroPET Imaging of Riboflavin Transporter 3 Expression in Myocardial Infarction/Reperfusion Rat Models with Radiofluorinated Riboflavin. ACS Pharmacol Transl Sci 2024; 7:2350-2357. [PMID: 39144563 PMCID: PMC11320726 DOI: 10.1021/acsptsci.4c00175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 08/16/2024]
Abstract
Riboflavin transporter 3 (RFVT3) represents a potential cardioprotective biotarget in energetic metabolism reprogramming after myocardial infarction/reperfusion (MI/R). This study investigated the feasibility of noninvasive real-time quantification of RFVT3 expression after MI/R with an radiolabeled probe 18F-RFTA in a preclinical rat model of MI/R. The tracer 18F-RFTA was radio-synthesized manually and characterized on the subjects of radiolabeling yield, radiochemical purity, and stability in vivo. MI/R and sham-operated rat models were confirmed by cardiac magnetic resonance imaging (cMRI) and single-photon-emission computed tomography (SPECT) myocardial perfusion imaging (MPI) with technetium-99m sestamibi (99mTc-MIBI). Positron emission tomography (PET) imaging of MI/R and sham-operated rat models were conducted with 18F-RFTA. Ex vivo autoradiography and RFVT3 immunohistochemical (IHC) staining were conducted to verify the RFVT3 expression in infarcted and normal myocardium. 18F-RFTA injection was prepared with high radiochemical purity (>95%) and kept stable in vitro and in vivo. 18F-RFTA PET revealed significant uptake in the infarcted myocardium at 8 h after reperfusion, as confirmed by lower 99mTc-MIBI perfusion and decreased intensity of cMRI. Conversely, there were only the tiniest uptakes in the normal myocardium and blocked infarcted myocardium, which was further corroborated by ex vivo autoradiography. The RFVT3 expression was further confirmed by IHC staining in the infarcted and normal myocardium. We first demonstrate the feasibility of imaging RFVT3 in infarcted myocardium. 18F-RFTA is an encouraging PET probe for imaging cardioprotective biotarget RFVT3 in mitochondrial energetic metabolism reprogramming after myocardial infarction. Noninvasive imaging of cardioprotective biotarget RFVT3 has potential value in the diagnosis and therapy of patients with MI.
Collapse
Affiliation(s)
- Jindian Li
- Department
of Nuclear Medicine, Fudan University Shanghai Cancer Center, Department
of Oncology, Shanghai Medical College, Fudan
University, Shanghai 200032, China
- Center
for Molecular Imaging and Translational Medicine, State Key Laboratory
of Molecular Vaccinology and Molecular Diagnostics, School of Public
Health, Xiamen University, Xiamen 361102, China
| | - Xingfang Hong
- Laboratory
of Pathogen Biology, School of Basic Medical Sciences, Dali University, Dali 671000, China
| | - Yingxi Chen
- Center
for Molecular Imaging and Translational Medicine, State Key Laboratory
of Molecular Vaccinology and Molecular Diagnostics, School of Public
Health, Xiamen University, Xiamen 361102, China
| | - Bin Yin
- The
First Affiliated Hospital, Center of Burn & Plastic and Wound
Healing Surgery, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Hongzhang Yang
- Center
for Molecular Imaging and Translational Medicine, State Key Laboratory
of Molecular Vaccinology and Molecular Diagnostics, School of Public
Health, Xiamen University, Xiamen 361102, China
| | - Changrong Shi
- Center
for Molecular Imaging and Translational Medicine, State Key Laboratory
of Molecular Vaccinology and Molecular Diagnostics, School of Public
Health, Xiamen University, Xiamen 361102, China
| | - Xinying Zeng
- Center
for Molecular Imaging and Translational Medicine, State Key Laboratory
of Molecular Vaccinology and Molecular Diagnostics, School of Public
Health, Xiamen University, Xiamen 361102, China
| | - Deliang Zhang
- Department
of Nuclear Medicine, Xiang’an Hospital
Affiliated to Xiamen University, Xiamen 361102, China
| | - Zhide Guo
- Center
for Molecular Imaging and Translational Medicine, State Key Laboratory
of Molecular Vaccinology and Molecular Diagnostics, School of Public
Health, Xiamen University, Xiamen 361102, China
| | - Xianzhong Zhang
- Department
of Nuclear Medicine, Peking Union Medical College Hospital & Theranostics
and Translational Research Center, National Infrastructures for Translational
Medicine, Institute of Clinical Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
4
|
Wu P, Xu L, Wang Q, Ma X, Wang X, Wang H, He S, Ru H, Zhao Y, Xiao Y, Zhang J, Wang X, An S, Hacker M, Li X, Zhang X, Wang Y, Yang M, Wu Z, Li S. Left Ventricular Remodelling Associated with the Transient Elevated [ 68Ga]Ga-Pentixafor Activity in the Remote Myocardium Following Acute Myocardial Infarction. Mol Imaging Biol 2024; 26:693-703. [PMID: 38641708 DOI: 10.1007/s11307-024-01912-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 02/17/2024] [Accepted: 03/14/2024] [Indexed: 04/21/2024]
Abstract
BACKGROUND Previous studies have initially reported accompanying elevated 2-deoxy-2[18F]fluoro-D-glucose ([18F]F-FDG) inflammatory activity in the remote area and its prognostic value after acute myocardial infarction (AMI). Non-invasive characterization of the accompanying inflammation in the remote myocardium may be of potency in guiding future targeted theranostics. [68Ga]Ga-Pentixafor targeting chemokine receptor 4 (CXCR4) on the surface of inflammatory cells is currently one of the promising inflammatory imaging agents. In this study, we sought to focus on the longitudinal evolution of [68Ga]Ga-Pentixafor activities in the remote myocardium following AMI and its association with cardiac function. METHODS Twelve AMI rats and six Sham rats serially underwent [68Ga]Ga-Pentixafor imaging at pre-operation, and 5, 7, 14 days post-operation. Maximum and mean standard uptake value (SUV) and target-to-background ratio (TBR) were assessed to indicate the uptake intensity. Gated [18F]F-FDG imaging and immunofluorescent staining were performed to obtain cardiac function and responses of pro-inflammatory and reparative macrophages, respectively. RESULTS The uptake of [68Ga]Ga-Pentixafor in the infarcted myocardium peaked at day 5 (all P = 0.003), retained at day 7 (all P = 0.011), and recovered at day 14 after AMI (P > 0.05), paralleling with the rise-fall pro-inflammatory M1 macrophages (P < 0.05). Correlated with the peak activity in the infarct territory, [68Ga]Ga-Pentixafor uptake in the remote myocardium on day 5 early after AMI significantly increased (AMI vs. Sham: SUVmean, SUVmax, and TBRmean: all P < 0.05), and strongly correlated with contemporaneous EDV and/or ESV (SUVmean and TBRmean: both P < 0.05). The transitory remote activity recovered as of day 7 post-AMI (AMI vs. Sham: P > 0.05). CONCLUSIONS Corresponding with the peaked [68Ga]Ga-Pentixafor activity in the infarcted myocardium, the activity in the remote region elevated accordingly and led to contemporaneous left ventricular remodelling early after AMI. Further studies are warranted to clarify its clinical application potential.
Collapse
Affiliation(s)
- Ping Wu
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, China
| | - Li Xu
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, China
| | - Qi Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Shanxi Medical University, Taiyuan, China
| | - Xiaofang Ma
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China
| | - Xinzhu Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Shanxi Medical University, Taiyuan, China
| | - Hongliang Wang
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, China
| | - Sheng He
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Huibin Ru
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, China
| | - Yuting Zhao
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China
| | - Yuxin Xiao
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China
| | - Jingying Zhang
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China
| | - Xinchao Wang
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, China
| | - Shaohui An
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, China
- Shanghai United Imaging Healthcare Co., Ltd., Shanghai, China
| | - Marcus Hacker
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Xiang Li
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Xiaoli Zhang
- Laboratory for Molecular Imaging, Department of Nuclear Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yuetao Wang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Minfu Yang
- Department of Nuclear Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Zhifang Wu
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, China
| | - Sijin Li
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China.
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
5
|
Lucinian YA, Martineau P, Abikhzer G, Harel F, Pelletier-Galarneau M. Novel tracers to assess myocardial inflammation with radionuclide imaging. J Nucl Cardiol 2024:102012. [PMID: 39069249 DOI: 10.1016/j.nuclcard.2024.102012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 07/08/2024] [Accepted: 07/15/2024] [Indexed: 07/30/2024]
Abstract
Myocardial inflammation plays a central role in the pathophysiology of various cardiac diseases. While FDG-PET is currently the primary method for molecular imaging of myocardial inflammation, its effectiveness is hindered by physiological myocardial uptake as well as its propensity for uptake by multiple disease-specific mechanisms. Novel radiotracers targeting diverse inflammatory immune cells and molecular pathways may provide unique insight through the visualization of underlying mechanisms central to the pathogenesis of inflammatory cardiac diseases, offering opportunities for increased understanding of immunocardiology. Moreover, the potentially enhanced specificity may lead to better quantification of disease activity, aiding in the guidance and monitoring of immunomodulatory therapy. This review aims to provide an update on advancements in non-FDG radiotracers for imaging myocardial inflammatory diseases, with a focus on cardiac sarcoidosis, myocarditis, and acute myocardial infarction.
Collapse
Affiliation(s)
| | | | - Gad Abikhzer
- Jewish General Hospital, Montreal, Quebec, Canada
| | | | | |
Collapse
|
6
|
Hilgendorf I, Frantz S, Frangogiannis NG. Repair of the Infarcted Heart: Cellular Effectors, Molecular Mechanisms and Therapeutic Opportunities. Circ Res 2024; 134:1718-1751. [PMID: 38843294 PMCID: PMC11164543 DOI: 10.1161/circresaha.124.323658] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 05/08/2024] [Indexed: 06/12/2024]
Abstract
The adult mammalian heart has limited endogenous regenerative capacity and heals through the activation of inflammatory and fibrogenic cascades that ultimately result in the formation of a scar. After infarction, massive cardiomyocyte death releases a broad range of damage-associated molecular patterns that initiate both myocardial and systemic inflammatory responses. TLRs (toll-like receptors) and NLRs (NOD-like receptors) recognize damage-associated molecular patterns (DAMPs) and transduce downstream proinflammatory signals, leading to upregulation of cytokines (such as interleukin-1, TNF-α [tumor necrosis factor-α], and interleukin-6) and chemokines (such as CCL2 [CC chemokine ligand 2]) and recruitment of neutrophils, monocytes, and lymphocytes. Expansion and diversification of cardiac macrophages in the infarcted heart play a major role in the clearance of the infarct from dead cells and the subsequent stimulation of reparative pathways. Efferocytosis triggers the induction and release of anti-inflammatory mediators that restrain the inflammatory reaction and set the stage for the activation of reparative fibroblasts and vascular cells. Growth factor-mediated pathways, neurohumoral cascades, and matricellular proteins deposited in the provisional matrix stimulate fibroblast activation and proliferation and myofibroblast conversion. Deposition of a well-organized collagen-based extracellular matrix network protects the heart from catastrophic rupture and attenuates ventricular dilation. Scar maturation requires stimulation of endogenous signals that inhibit fibroblast activity and prevent excessive fibrosis. Moreover, in the mature scar, infarct neovessels acquire a mural cell coat that contributes to the stabilization of the microvascular network. Excessive, prolonged, or dysregulated inflammatory or fibrogenic cascades accentuate adverse remodeling and dysfunction. Moreover, inflammatory leukocytes and fibroblasts can contribute to arrhythmogenesis. Inflammatory and fibrogenic pathways may be promising therapeutic targets to attenuate heart failure progression and inhibit arrhythmia generation in patients surviving myocardial infarction.
Collapse
Affiliation(s)
- Ingo Hilgendorf
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine at the University of Freiburg, Freiburg, Germany
| | - Stefan Frantz
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx NY
| |
Collapse
|
7
|
Callegari S, Feher A, Smolderen KG, Mena-Hurtado C, Sinusas AJ. Multi-modality imaging for assessment of the microcirculation in peripheral artery disease: Bench to clinical practice. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2024; 42:100400. [PMID: 38779485 PMCID: PMC11108852 DOI: 10.1016/j.ahjo.2024.100400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 05/07/2024] [Indexed: 05/25/2024]
Abstract
Peripheral artery disease (PAD) is a highly prevalent disorder with a high risk of mortality and amputation despite the introduction of novel medical and procedural treatments. Microvascular disease (MVD) is common among patients with PAD, and despite the established role as a predictor of amputations and mortality, MVD is not routinely assessed as part of current standard practice. Recent pre-clinical and clinical perfusion and molecular imaging studies have confirmed the important role of MVD in the pathogenesis and outcomes of PAD. The recent advancements in the imaging of the peripheral microcirculation could lead to a better understanding of the pathophysiology of PAD, and result in improved risk stratification, and our evaluation of response to therapies. In this review, we will discuss the current understanding of the anatomy and physiology of peripheral microcirculation, and the role of imaging for assessment of perfusion in PAD, and the latest advancements in molecular imaging. By highlighting the latest advancements in multi-modality imaging of the peripheral microcirculation, we aim to underscore the most promising imaging approaches and highlight potential research opportunities, with the goal of translating these approaches for improved and personalized management of PAD in the future.
Collapse
Affiliation(s)
- Santiago Callegari
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, USA
- Vascular Medicine Outcomes Program, Yale University, New Haven, CT, USA
| | - Attila Feher
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, USA
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Kim G. Smolderen
- Vascular Medicine Outcomes Program, Yale University, New Haven, CT, USA
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Carlos Mena-Hurtado
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, USA
- Vascular Medicine Outcomes Program, Yale University, New Haven, CT, USA
| | - Albert J. Sinusas
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, USA
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| |
Collapse
|
8
|
Toner YC, Prévot G, van Leent MMT, Munitz J, Oosterwijk R, Verschuur AVD, van Elsas Y, Peric V, Maas RJF, Ranzenigo A, Morla-Folch J, Wang W, Umali M, de Dreu A, Fernandes JC, Sullivan NAT, Maier A, Mason C, Reiner T, Fayad ZA, Mulder WJM, Teunissen AJP, Pérez-Medina C. Macrophage PET imaging in mouse models of cardiovascular disease and cancer with an apolipoprotein-inspired radiotracer. NPJ IMAGING 2024; 2:12. [PMID: 38765879 PMCID: PMC11096117 DOI: 10.1038/s44303-024-00009-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 02/06/2024] [Indexed: 05/22/2024]
Abstract
Macrophages are key inflammatory mediators in many pathological conditions, including cardiovascular disease (CVD) and cancer, the leading causes of morbidity and mortality worldwide. This makes macrophage burden a valuable diagnostic marker and several strategies to monitor these cells have been reported. However, such strategies are often high-priced, non-specific, invasive, and/or not quantitative. Here, we developed a positron emission tomography (PET) radiotracer based on apolipoprotein A1 (ApoA1), the main protein component of high-density lipoprotein (HDL), which has an inherent affinity for macrophages. We radiolabeled an ApoA1-mimetic peptide (mA1) with zirconium-89 (89Zr) to generate a lipoprotein-avid PET probe (89Zr-mA1). We first characterized 89Zr-mA1's affinity for lipoproteins in vitro by size exclusion chromatography. To study 89Zr-mA1's in vivo behavior and interaction with endogenous lipoproteins, we performed extensive studies in wildtype C57BL/6 and Apoe-/- hypercholesterolemic mice. Subsequently, we used in vivo PET imaging to study macrophages in melanoma and myocardial infarction using mouse models. The tracer's cell specificity was assessed by histology and mass cytometry (CyTOF). Our data show that 89Zr-mA1 associates with lipoproteins in vitro. This is in line with our in vivo experiments, in which we observed longer 89Zr-mA1 circulation times in hypercholesterolemic mice compared to C57BL/6 controls. 89Zr-mA1 displayed a tissue distribution profile similar to ApoA1 and HDL, with high kidney and liver uptake as well as substantial signal in the bone marrow and spleen. The tracer also accumulated in tumors of melanoma-bearing mice and in the ischemic myocardium of infarcted animals. In these sites, CyTOF analyses revealed that natZr-mA1 was predominantly taken up by macrophages. Our results demonstrate that 89Zr-mA1 associates with lipoproteins and hence accumulates in macrophages in vivo. 89Zr-mA1's high uptake in these cells makes it a promising radiotracer for non-invasively and quantitatively studying conditions characterized by marked changes in macrophage burden.
Collapse
Affiliation(s)
- Yohana C. Toner
- 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
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Geoffrey Prévot
- 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
| | - Jazz Munitz
- 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
| | - Roderick Oosterwijk
- 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
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Anna Vera D. Verschuur
- 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
| | - Yuri van Elsas
- 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
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Vedran Peric
- 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
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Rianne J. F. Maas
- 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
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Anna Ranzenigo
- 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
| | - Judit Morla-Folch
- 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
| | - William Wang
- 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
| | - Martin Umali
- 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
| | - Anne de Dreu
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Jessica Chimene Fernandes
- 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
| | - Nathaniel A. T. Sullivan
- 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
| | - Alexander Maier
- 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
- Department of Cardiology and Angiology, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christian Mason
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY USA
- Department of Radiology, Weill Cornell Medical College, New York, NY USA
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 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
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Willem J. M. Mulder
- 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
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - 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
| | - Carlos Pérez-Medina
- 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
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| |
Collapse
|
9
|
Strunk M, Heo GS, Hess A, Luehmann H, Ross TL, Gropler RJ, Bengel FM, Liu Y, Thackeray JT. Toward Quantitative Multisite Preclinical Imaging Studies in Acute Myocardial Infarction: Evaluation of the Immune-Fibrosis Axis. J Nucl Med 2024; 65:287-293. [PMID: 38176717 DOI: 10.2967/jnumed.123.266526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 11/09/2023] [Indexed: 01/06/2024] Open
Abstract
The immune-fibrosis axis plays a critical role in cardiac remodeling after acute myocardial infarction. Imaging approaches to monitor temporal inflammation and fibroblast activation in mice have seen wide application in recent years. However, the repeatability of quantitative measurements remains challenging, particularly across multiple imaging centers. We aimed to determine reproducibility of quantitative inflammation and fibroblast activation images acquired at 2 facilities after myocardial infarction in mice. Methods: Mice underwent coronary artery ligation and sequential imaging with 68Ga-DOTA-ECL1i to assess chemokine receptor type 2 expression at 3 d after myocardial infarction and 68Ga-FAPI-46 to assess fibroblast activation protein expression at 7 d after myocardial infarction. Images were acquired at 1 center using either a local or a consensus protocol developed with the second center; the protocols differed in the duration of isoflurane anesthesia and the injected tracer dose. A second group of animals were scanned at the second site using the consensus protocol. Image analyses performed by each site and just by 1 site were also compared. Results: The uptake of 68Ga-DOTA-ECL1i in the infarct territory tended to be higher when the consensus protocol was used (P = 0.03). No difference was observed between protocol acquisitions for 68Ga-FAPI-46. Compared with the local protocol, the consensus protocol decreased variability between individual animals. When a matched consensus protocol was used, the 68Ga-DOTA-ECL1i infarct territory percentage injected dose per gram of tissue was higher on images acquired at site B than on those acquired at site A (P = 0.006). When normalized to body weight as SUV, this difference was mitigated. Both the percentage injected dose per gram of tissue and the SUV were comparable between sites for 68Ga-FAPI-46. Image analyses at the sites differed significantly, but this difference was mitigated when all images were analyzed at site A. Conclusion: The application of a standardized acquisition protocol may lower variability within datasets and facilitate comparison of molecular radiotracer distribution between preclinical imaging centers. Like clinical studies, multicenter preclinical studies should use centralized core-based image analysis to maximize reproducibility across sites.
Collapse
Affiliation(s)
- Maja Strunk
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany; and
| | - Gyu Seong Heo
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Annika Hess
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany; and
| | - Hannah Luehmann
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Tobias L Ross
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany; and
| | - Robert J Gropler
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany; and
| | - Yongjian Liu
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - James T Thackeray
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany; and
| |
Collapse
|
10
|
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
|
11
|
Konrad M, Rinscheid A, Wienand G, Nittbaur B, Wester HJ, Janzen T, Lapa C, Pfob CH, Schottelius M. [ 99mTc]Tc-PentixaTec: development, extensive pre-clinical evaluation, and first human experience. Eur J Nucl Med Mol Imaging 2023; 50:3937-3948. [PMID: 37597009 PMCID: PMC10611619 DOI: 10.1007/s00259-023-06395-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 08/05/2023] [Indexed: 08/21/2023]
Abstract
PURPOSE The clinical success non-invasive imaging of CXCR4 expression using [68 Ga]Ga-PentixaFor-PET warrants an expansion of the targeting concept towards conventional scintigraphy/SPECT with their lower cost and general availability. To this aim, we developed and comparatively evaluated a series of 99mTc-labeled cyclic pentapeptides based on the PentixaFor scaffold. METHODS Six mas3-conjugated CPCR4 analogs with different 4-aminobenzoic acid (Abz)-D-Ala-D-Arg-aa3 linkers (L1-L6) as well as the corresponding HYNIC- and N4-analogs of L6-CPCR4 were synthesized via standard SPPS. Competitive binding studies (IC50 and IC50inv) were carried out using Jurkat T cell lymphoma cells and [125I]FC-131 as radioligand. Internalization kinetics were investigated using hCXCR4-overexpressing Chem-1 cells. Biodistribution studies and small animal SPECT/CT imaging (1 h p.i.) were carried out using Jurkat xenograft bearing CB17/SCID mice. Based on the preclinical results, [99mTc]Tc-N4-L6-CPCR4 ([99mTc]Tc-PentixaTec) was selected for an early translation to the human setting. Five patients with hematologic malignancies underwent [99mTc]Tc-N4-L6-CPCR4 SPECT/planar imaging with individual dosimetry. RESULTS Of the six mas3-conjugated peptides, mas3-L6-CPCR4 (mas3-dap-r-a-Abz-CPCR4) showed the highest CXCR4 affinity (IC50 = 5.0 ± 1.3 nM). Conjugation with N4 (N4-L6-CPCR4) further improved hCXCR4 affinity to 0.6 ± 0.1 nM. [99mTc]Tc-N4-L6-CPCR4 also showed the most efficient internalization (97% of total cellular activity at 2 h) and the highest tumor accumulation (8.6 ± 1.3% iD/g, 1 h p.i.) of the compounds investigated. Therefore, [99mTc]Tc-N4-L6-CPCR4 (termed [99mTc]Tc-PentixaTec) was selected for first-in-human application. [99mTc]Tc-PentixaTec was well tolerated, exhibits a favorable biodistribution and dosimetry profile (2.1-3.4 mSv per 500 MBq) and excellent tumor/background ratios in SPECT and planar imaging. CONCLUSION The successive optimization of the amino acid composition of the linker structure and the N-terminal 99mTc-labeling strategies (mas3 vs HYNIC vs N4) has provided [99mTc]Tc-PentixaTec as a novel, highly promising CXCR4-targeted SPECT agent for clinical application. With its excellent CXCR4 affinity, efficient internalization, high uptake in CXCR4-expressing tissues, suitable clearance/biodistribution characteristics, and favorable human dosimetry, it holds great potential for further clinical use.
Collapse
Affiliation(s)
- Matthias Konrad
- Chair for Pharmaceutical Radiochemistry, Faculties of Chemistry and Medicine, Technische Universität München, 85748, Garching, Germany
| | - Andreas Rinscheid
- Medical Physics and Radiation Protection, University Hospital Augsburg, Stenglinstrasse 2, 86156, Augsburg, Germany
| | - Georgine Wienand
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Stenglinstrasse 2, 86156, Augsburg, Germany
| | - Bernd Nittbaur
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Stenglinstrasse 2, 86156, Augsburg, Germany
| | - Hans-Jürgen Wester
- Chair for Pharmaceutical Radiochemistry, Faculties of Chemistry and Medicine, Technische Universität München, 85748, Garching, Germany
| | - Tilman Janzen
- Medical Physics and Radiation Protection, University Hospital Augsburg, Stenglinstrasse 2, 86156, Augsburg, Germany
| | - Constantin Lapa
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Stenglinstrasse 2, 86156, Augsburg, Germany
| | - Christian Helmut Pfob
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Stenglinstrasse 2, 86156, Augsburg, Germany.
| | - Margret Schottelius
- Translational Radiopharmaceutical Sciences, Department of Nuclear Medicine and Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Rue du Bugnon 25A, Agora, CH-1011, Lausanne, Switzerland.
- AGORA, Pôle de Recherche Sur Le Cancer, 1011, Lausanne, Switzerland.
- SCCL Swiss Cancer Center Leman, 1011, Lausanne, Switzerland.
| |
Collapse
|
12
|
Kersting D, Mavroeidi IA, Settelmeier S, Seifert R, Schuler M, Herrmann K, Rassaf T, Rischpler C. Molecular Imaging Biomarkers in Cardiooncology: A View on Established Technologies and Future Perspectives. J Nucl Med 2023; 64:29S-38S. [PMID: 37918843 DOI: 10.2967/jnumed.122.264868] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 09/05/2023] [Indexed: 11/04/2023] Open
Abstract
Novel therapeutic options have significantly improved survival and long-term outcomes in many cancer entities. Unfortunately, this improvement in outcome is often accompanied by new and increasingly relevant therapy-related cardiovascular toxicity. In this context, cardiooncology has emerged as a new field of interdisciplinary individual patient care. Important tasks are pretherapeutic risk stratification and early detection and treatment of cardiotoxicity, which comprises cardiac damage in relation to cardiovascular comorbidities, the tumor disease, and cancer treatment. Clinical manifestations can cover a broad spectrum, ranging from subtle and usually asymptomatic abnormalities to serious acute or chronic complications. Typical manifestations include acute and chronic heart failure, myo- and pericarditis, arrythmias, ischemia, and endothelial damage. They can be related to almost all current cancer treatments, including cytotoxic chemotherapy, targeted therapy, immunotherapy, hormonal therapy, and radiotherapy. Molecular imaging biomarkers can aid in pretherapeutic cardiooncologic assessment for primary prevention and personalized surveillance, detection, and differential diagnosis of cardiotoxic complications. Potential advantages over conventional diagnostics are the higher detection sensitivity for subtle changes in cardiac homeostasis, higher reproducibility, and better observer independence. Hybrid imaging with highly sensitive PET/MRI may be particularly suited for early diagnosis. Important technologies that are encouraged in current multidisciplinary guidelines are equilibrium radionuclide angiography for evaluation of ventricular function and chamber morphology, as well as myocardial perfusion imaging for additional detection of ischemia. Novel modalities that may detect even earlier signs of cardiotoxicity comprise 123I-metaiodobenzylguanidine SPECT to visualize sympathetic innervation, 18F-FDG and somatostatin receptor (68Ga-DOTATOC/DOTATATE) PET to indicate a metabolic shift and inflammation, and 68Ga-fibroblast activation protein inhibitor PET to monitor cardiac remodeling. In addition, PET imaging of mitochondrial function has recently been introduced in preclinical models and will potentially broaden the field of application through higher sensitivity and specificity and by enabling higher individualization of diagnostic concepts.
Collapse
Affiliation(s)
- David Kersting
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany;
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| | - Ilektra-Antonia Mavroeidi
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany; and
| | - Stephan Settelmeier
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Robert Seifert
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| | - Martin Schuler
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany; and
| | - Ken Herrmann
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Christoph Rischpler
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| |
Collapse
|
13
|
Renard I, Domarkas J, Poty S, Burke BP, Roberts DP, Goze C, Denat F, Cawthorne CJ, Archibald SJ. In vivo validation of 68Ga-labeled AMD3100 conjugates for PET imaging of CXCR4. Nucl Med Biol 2023; 120-121:108335. [PMID: 37068392 DOI: 10.1016/j.nucmedbio.2023.108335] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 03/13/2023] [Accepted: 03/21/2023] [Indexed: 04/19/2023]
Abstract
INTRODUCTION The chemokine receptor CXCR4 has been shown to be over-expressed in multiple types of cancer and is usually associated with aggressive phenotypes and poor prognosis. Successfully targeting and imaging the expression level of this receptor in tumours could inform treatment selection and facilitate patient stratification. METHODS Known conjugates of AMD3100 that are specific to CXCR4 have been radiolabelled with gallium-68 and evaluated in naïve and tumour-bearing mice. Tumour uptake of the radiotracers was compared to the known CXCR4-specific PET imaging agent, [68Ga]Pentixafor. RESULTS Ex vivo biodistribution in naïve animals showed CXCR4-mediated uptake in the liver with both radiotracers, confirmed by blocking experiments with the high affinity CXCR4 antagonist Cu2CB-Bicyclam (IC50 = 3 nM). PET/CT imaging studies revealed one tracer to have a higher accumulation in the tumour (SUVMean of 0.89 ± 0.14 vs 0.32 ± 0.11). CXCR4-specificity of the best performing tracer was confirmed by administration of a blocking dose of Cu2CB-Bicyclam, showing a 3- and 6-fold decrease in tumour and liver uptake, respectively. CONCLUSION AND ADVANCES IN KNOWLEDGE This initial study offers some interesting insights on the impact of some structural features on the pharmacokinetics and metabolic stability of the radiotracer. Additionally, as Pentixafor only binds to human CXCR4, the development of CXCR4-targeted imaging agents that bind to the receptor across different species could significantly help with preclinical evaluation of new CXCR4-specific therapeutics.
Collapse
Affiliation(s)
- Isaline Renard
- Centre for Biomedicine and PET Research Centre, Hull York Medical School, University of Hull, Hull HU6 7RX, UK
| | - Juozas Domarkas
- Centre for Biomedicine and PET Research Centre, Hull York Medical School, University of Hull, Hull HU6 7RX, UK
| | - Sophie Poty
- Institut de Chimie Moléculaire de l'Université de Bourgogne, UMR 6302, CNRS, Université Bourgogne Franche-Comté, Dijon, France
| | - Benjamin P Burke
- Centre for Biomedicine and PET Research Centre, Hull York Medical School, University of Hull, Hull HU6 7RX, UK
| | - David P Roberts
- Centre for Biomedicine and PET Research Centre, Hull York Medical School, University of Hull, Hull HU6 7RX, UK
| | - Christine Goze
- Institut de Chimie Moléculaire de l'Université de Bourgogne, UMR 6302, CNRS, Université Bourgogne Franche-Comté, Dijon, France.
| | - Franck Denat
- Institut de Chimie Moléculaire de l'Université de Bourgogne, UMR 6302, CNRS, Université Bourgogne Franche-Comté, Dijon, France.
| | - Christopher J Cawthorne
- Centre for Biomedicine and PET Research Centre, Hull York Medical School, University of Hull, Hull HU6 7RX, UK; Nuclear Medicine & Molecular Imaging, Department of Imaging & Pathology, KU Leuven, 3000 Leuven, Belgium.
| | - Stephen J Archibald
- Centre for Biomedicine and PET Research Centre, Hull York Medical School, University of Hull, Hull HU6 7RX, UK.
| |
Collapse
|
14
|
Senders ML, Calcagno C, Tawakol A, Nahrendorf M, Mulder WJM, Fayad ZA. PET/MR imaging of inflammation in atherosclerosis. Nat Biomed Eng 2023; 7:202-220. [PMID: 36522465 DOI: 10.1038/s41551-022-00970-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 10/25/2022] [Indexed: 12/23/2022]
Abstract
Myocardial infarction, stroke, mental disorders, neurodegenerative processes, autoimmune diseases, cancer and the human immunodeficiency virus impact the haematopoietic system, which through immunity and inflammation may aggravate pre-existing atherosclerosis. The interplay between the haematopoietic system and its modulation of atherosclerosis has been studied by imaging the cardiovascular system and the activation of haematopoietic organs via scanners integrating positron emission tomography and resonance imaging (PET/MRI). In this Perspective, we review the applicability of integrated whole-body PET/MRI for the study of immune-mediated phenomena associated with haematopoietic activity and cardiovascular disease, and discuss the translational opportunities and challenges of the technology.
Collapse
Affiliation(s)
- Max L Senders
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Claudia Calcagno
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ahmed Tawakol
- Cardiology Division and Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Matthias Nahrendorf
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Willem J M Mulder
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands.
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS) and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands.
- Laboratory of Chemical Biology, Department of Biochemical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands.
| | - Zahi A Fayad
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
15
|
Gunata M, Parlakpinar H. Experimental heart failure models in small animals. Heart Fail Rev 2023; 28:533-554. [PMID: 36504404 DOI: 10.1007/s10741-022-10286-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/08/2022] [Indexed: 12/14/2022]
Abstract
Heart failure (HF) is one of the most critical health and economic burdens worldwide, and its prevalence is continuously increasing. HF is a disease that occurs due to a pathological change arising from the function or structure of the heart tissue and usually progresses. Numerous experimental HF models have been created to elucidate the pathophysiological mechanisms that cause HF. An understanding of the pathophysiology of HF is essential for the development of novel efficient therapies. During the past few decades, animal models have provided new insights into the complex pathogenesis of HF. Success in the pathophysiology and treatment of HF has been achieved by using animal models of HF. The development of new in vivo models is critical for evaluating treatments such as gene therapy, mechanical devices, and new surgical approaches. However, each animal model has advantages and limitations, and none of these models is suitable for studying all aspects of HF. Therefore, the researchers have to choose an appropriate experimental model that will fully reflect HF. Despite some limitations, these animal models provided a significant advance in the etiology and pathogenesis of HF. Also, experimental HF models have led to the development of new treatments. In this review, we discussed widely used experimental HF models that continue to provide critical information for HF patients and facilitate the development of new treatment strategies.
Collapse
Affiliation(s)
- Mehmet Gunata
- Department of Medical Pharmacology, Faculty of Medicine, Inonu University, Malatya, 44280, Türkiye
| | - Hakan Parlakpinar
- Department of Medical Pharmacology, Faculty of Medicine, Inonu University, Malatya, 44280, Türkiye.
| |
Collapse
|
16
|
Zhang RYK, Cochran BJ, Thomas SR, Rye KA. Impact of Reperfusion on Temporal Immune Cell Dynamics After Myocardial Infarction. J Am Heart Assoc 2023; 12:e027600. [PMID: 36789837 PMCID: PMC10111498 DOI: 10.1161/jaha.122.027600] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Excessive inflammation and impaired healing of cardiac tissue following a myocardial infarction (MI) can drive the development of heart failure. Cardiac repair begins immediately after the onset of MI and continues for months. The repair process can be divided into the following 3 overlapping phases, each having distinct functions and sequelae: the inflammatory phase, the proliferative phase, and the maturation phase. Macrophages, neutrophils, and lymphocytes are present in the myocardium throughout the repair process and govern the duration and function of each of these phases. However, changes in the functions of these cell types across each phase are poorly characterized. Numerous immunomodulatory therapies that specifically target inflammation have been developed for promoting cardiac repair and preventing heart failure after MI. However, these treatments have been largely unsuccessful in large-scale clinical randomized controlled trials. A potential explanation for this failure is the lack of a thorough understanding of the time-dependent evolution of the functions of immune cells after a major cardiovascular event. Failure to account for this temporal plasticity in cell function may reduce the efficacy of immunomodulatory approaches that target cardiac repair. This review is concerned with how the functions of different immune cells change with time following an MI. Improved understanding of the temporal changes in immune cell function is important for the future development of effective and targeted treatments for preventing heart failure after MI.
Collapse
Affiliation(s)
| | - Blake J Cochran
- School of Medical Sciences University of New South Wales Sydney New South Wales
| | - Shane R Thomas
- School of Medical Sciences University of New South Wales Sydney New South Wales
| | - Kerry-Anne Rye
- School of Medical Sciences University of New South Wales Sydney New South Wales
| |
Collapse
|
17
|
Bouvain P, Ding Z, Kadir S, Kleimann P, Kluge N, Tiren ZB, Steckel B, Flocke V, Zalfen R, Petzsch P, Wachtmeister T, John G, Subramaniam N, Krämer W, Strasdeit T, Mehrabipour M, Moll JM, Schubert R, Ahmadian MR, Bönner F, Boeken U, Westenfeld R, Engel DR, Kelm M, Schrader J, Köhrer K, Grandoch M, Temme S, Flögel U. Non-invasive mapping of systemic neutrophil dynamics upon cardiovascular injury. NATURE CARDIOVASCULAR RESEARCH 2023; 2:126-143. [PMID: 39196054 PMCID: PMC11357992 DOI: 10.1038/s44161-022-00210-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 12/22/2022] [Indexed: 08/29/2024]
Abstract
Neutrophils play a complex role during onset of tissue injury and subsequent resolution and healing. To assess neutrophil dynamics upon cardiovascular injury, here we develop a non-invasive, background-free approach for specific mapping of neutrophil dynamics by whole-body magnetic resonance imaging using targeted multimodal fluorine-loaded nanotracers engineered with binding peptides specifically directed against murine or human neutrophils. Intravenous tracer application before injury allowed non-invasive three-dimensional visualization of neutrophils within their different hematopoietic niches over the entire body and subsequent monitoring of their egress into affected tissues. Stimulated murine and human neutrophils exhibited enhanced labeling due to upregulation of their target receptors, which could be exploited as an in vivo readout for their activation state in both sterile and nonsterile cardiovascular inflammation. This non-invasive approach will allow us to identify hidden origins of bacterial or sterile inflammation in patients and also to unravel cardiovascular disease states on the verge of severe aggravation due to enhanced neutrophil infiltration or activation.
Collapse
Affiliation(s)
- Pascal Bouvain
- Experimental Cardiovascular Imaging, Institute for Molecular Cardiology, Heinrich Heine University, Düsseldorf, Germany
| | - Zhaoping Ding
- Experimental Cardiovascular Imaging, Institute for Molecular Cardiology, Heinrich Heine University, Düsseldorf, Germany
| | - Shiwa Kadir
- Experimental Cardiovascular Imaging, Institute for Molecular Cardiology, Heinrich Heine University, Düsseldorf, Germany
| | - Patricia Kleimann
- Experimental Cardiovascular Imaging, Institute for Molecular Cardiology, Heinrich Heine University, Düsseldorf, Germany
| | - Nils Kluge
- Experimental Cardiovascular Imaging, Institute for Molecular Cardiology, Heinrich Heine University, Düsseldorf, Germany
| | - Zeynep-Büsra Tiren
- Experimental Cardiovascular Imaging, Institute for Molecular Cardiology, Heinrich Heine University, Düsseldorf, Germany
| | - Bodo Steckel
- Experimental Cardiovascular Imaging, Institute for Molecular Cardiology, Heinrich Heine University, Düsseldorf, Germany
| | - Vera Flocke
- Experimental Cardiovascular Imaging, Institute for Molecular Cardiology, Heinrich Heine University, Düsseldorf, Germany
| | - Ria Zalfen
- Experimental Cardiovascular Imaging, Institute for Molecular Cardiology, Heinrich Heine University, Düsseldorf, Germany
| | - Patrick Petzsch
- Biological and Medical Research Center (BMFZ), Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Thorsten Wachtmeister
- Biological and Medical Research Center (BMFZ), Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Gordon John
- Dental Office/Oral Surgery, Dr. G. John, Plauen, Germany
| | - Nirojah Subramaniam
- Institute for Experimental Immunology and Imaging, Department of Immunodynamics, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Wolfgang Krämer
- Department of Pharmaceutical Technology and Biopharmacy, Albert Ludwig University, Freiburg im Breisgau, Germany
| | - Tobias Strasdeit
- Institute of Neuro- and Sensory Physiology, Heinrich Heine University, Düsseldorf, Germany
| | - Mehrnaz Mehrabipour
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Jens M Moll
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Rolf Schubert
- Department of Pharmaceutical Technology and Biopharmacy, Albert Ludwig University, Freiburg im Breisgau, Germany
| | - Mohammad Reza Ahmadian
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Florian Bönner
- Department of Cardiology, Pneumology and Angiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Udo Boeken
- Clinic for Cardiac Surgery, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Ralf Westenfeld
- Department of Cardiology, Pneumology and Angiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Daniel Robert Engel
- Institute for Experimental Immunology and Imaging, Department of Immunodynamics, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Malte Kelm
- Department of Cardiology, Pneumology and Angiology, University Hospital Düsseldorf, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Heinrich Heine University, Düsseldorf, Germany
| | - Jürgen Schrader
- Experimental Cardiovascular Imaging, Institute for Molecular Cardiology, Heinrich Heine University, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Heinrich Heine University, Düsseldorf, Germany
| | - Karl Köhrer
- Biological and Medical Research Center (BMFZ), Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Maria Grandoch
- Institute for Translational Pharmacology, Heinrich Heine University, Düsseldorf, Germany
| | - Sebastian Temme
- Experimental Cardiovascular Imaging, Institute for Molecular Cardiology, Heinrich Heine University, Düsseldorf, Germany
- Department of Anesthesiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Ulrich Flögel
- Experimental Cardiovascular Imaging, Institute for Molecular Cardiology, Heinrich Heine University, Düsseldorf, Germany.
- Department of Cardiology, Pneumology and Angiology, University Hospital Düsseldorf, Düsseldorf, Germany.
- Cardiovascular Research Institute Düsseldorf (CARID), Heinrich Heine University, Düsseldorf, Germany.
| |
Collapse
|
18
|
Saba L, Loewe C, Weikert T, Williams MC, Galea N, Budde RPJ, Vliegenthart R, Velthuis BK, Francone M, Bremerich J, Natale L, Nikolaou K, Dacher JN, Peebles C, Caobelli F, Redheuil A, Dewey M, Kreitner KF, Salgado R. State-of-the-art CT and MR imaging and assessment of atherosclerotic carotid artery disease: standardization of scanning protocols and measurements-a consensus document by the European Society of Cardiovascular Radiology (ESCR). Eur Radiol 2023; 33:1063-1087. [PMID: 36194267 PMCID: PMC9889495 DOI: 10.1007/s00330-022-09024-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 06/26/2022] [Accepted: 06/30/2022] [Indexed: 02/04/2023]
Abstract
The European Society of Cardiovascular Radiology (ESCR) is the European specialist society of cardiac and vascular imaging. This society's highest priority is the continuous improvement, development, and standardization of education, training, and best medical practice, based on experience and evidence. The present intra-society consensus is based on the existing scientific evidence and on the individual experience of the members of the ESCR writing group on carotid diseases, the members of the ESCR guidelines committee, and the members of the executive committee of the ESCR. The recommendations published herein reflect the evidence-based society opinion of ESCR. We have produced a twin-papers consensus, indicated through the documents as respectively "Part I" and "Part II." The first document (Part I) begins with a discussion of features, role, indications, and evidence for CT and MR imaging-based diagnosis of carotid artery disease for risk stratification and prediction of stroke (Section I). It then provides an extensive overview and insight into imaging-derived biomarkers and their potential use in risk stratification (Section II). Finally, detailed recommendations about optimized imaging technique and imaging strategies are summarized (Section III). The second part of this consensus paper (Part II) is focused on structured reporting of carotid imaging studies with CT/MR. KEY POINTS: • CT and MR imaging-based evaluation of carotid artery disease provides essential information for risk stratification and prediction of stroke. • Imaging-derived biomarkers and their potential use in risk stratification are evolving; their correct interpretation and use in clinical practice must be well-understood. • A correct imaging strategy and scan protocol will produce the best possible results for disease evaluation.
Collapse
Affiliation(s)
- Luca Saba
- Department of Radiology, University of Cagliari, Cagliari, Italy
| | - Christian Loewe
- Division of Cardiovascular and Interventional Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Thomas Weikert
- Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Michelle C Williams
- BHF Centre for Cardiovascular Science, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH164SB, UK
- Edinburgh Imaging Facility QMRI, University of Edinburgh, Edinburgh, UK
| | - Nicola Galea
- Policlinico Umberto I, Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Rome, Italy
| | - Ricardo P J Budde
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Rozemarijn Vliegenthart
- Department of Radiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Birgitta K Velthuis
- Department of Radiology, Utrecht University Medical Center, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Marco Francone
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072, Milan, Italy
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Jens Bremerich
- Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Luigi Natale
- Department of Radiological Sciences - Institute of Radiology, Catholic University of Rome, "A. Gemelli" University Hospital, Rome, Italy
| | - Konstantin Nikolaou
- Department of Diagnostic and Interventional Radiology, University of Tuebingen, Tübingen, Germany
| | - Jean-Nicolas Dacher
- Department of Radiology, Normandie University, UNIROUEN, INSERM U1096 - Rouen University Hospital, F 76000, Rouen, France
| | - Charles Peebles
- Department of Cardiothoracic Radiology, University Hospital Southampton, Southampton, UK
| | - Federico Caobelli
- University Clinic of Nuclear Medicine Inselspital Bern, University of Bern, Bern, Switzerland
| | - Alban Redheuil
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
- Department of Cardiovascular and Thoracic, Imaging and Interventional Radiology, Institute of Cardiology, APHP, Pitié-Salpêtrière University Hospital, Paris, France
- Laboratoire d'Imagerie Biomédicale, Sorbonne Universités, UPMC Univ Paris 06, INSERM 1146, CNRS 7371, Paris, France
| | - Marc Dewey
- Department of Radiology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Karl-Friedrich Kreitner
- Department of Diagnostic and Interventional Radiology, University Medical Center, Mainz; Langenbeckstraße 1, 55131, Mainz, Germany
| | - Rodrigo Salgado
- Department of Radiology, Antwerp University Hospital & Antwerp University, Holy Heart Lier, Belgium.
| |
Collapse
|
19
|
Heo GS, Diekmann J, Thackeray JT, Liu Y. Nuclear Methods for Immune Cell Imaging: Bridging Molecular Imaging and Individualized Medicine. Circ Cardiovasc Imaging 2023; 16:e014067. [PMID: 36649445 PMCID: PMC9858352 DOI: 10.1161/circimaging.122.014067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Inflammation is a key mechanistic contributor to the progression of cardiovascular disease, from atherosclerosis through ischemic injury and overt heart failure. Recent evidence has identified specific roles of immune cell subpopulations in cardiac pathogenesis that diverges between individual patients. Nuclear imaging approaches facilitate noninvasive and serial quantification of inflammation severity, offering the opportunity to predict eventual outcome, stratify patient risk, and guide novel targeted molecular therapies against specific leukocyte subpopulations. Here, we will discuss the established and emerging nuclear imaging methods to label and track exogenous and endogenous immune cells, with a particular focus on clinical situations in which targeted molecular inflammation imaging would be advantageous. The expanding options for imaging inflammation provide the foundation to bridge between molecular imaging and individual therapy.
Collapse
Affiliation(s)
- Gyu Seong Heo
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO (G.S.H., Y. L.)
| | - Johanna Diekmann
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany (J.D., J.T.T.)
| | - James T Thackeray
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany (J.D., J.T.T.)
| | - Yongjian Liu
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO (G.S.H., Y. L.)
| |
Collapse
|
20
|
Li X, Wu M, Li J, Guo Q, Zhao Y, Zhang X. Advanced targeted nanomedicines for vulnerable atherosclerosis plaque imaging and their potential clinical implications. Front Pharmacol 2022; 13:906512. [PMID: 36313319 PMCID: PMC9606597 DOI: 10.3389/fphar.2022.906512] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 09/20/2022] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis plaques caused by cerebrovascular and coronary artery disease have been the leading cause of death and morbidity worldwide. Precise assessment of the degree of atherosclerotic plaque is critical for predicting the risk of atherosclerosis plaques and monitoring postinterventional outcomes. However, traditional imaging techniques to predict cardiocerebrovascular events mainly depend on quantifying the percentage reduction in luminal diameter, which would immensely underestimate non-stenotic high-risk plaque. Identifying the degree of atherosclerosis plaques still remains highly limited. vNanomedicine-based imaging techniques present unique advantages over conventional techniques due to the superior properties intrinsic to nanoscope, which possess enormous potential for characterization and detection of the features of atherosclerosis plaque vulnerability. Here, we review recent advancements in the development of targeted nanomedicine-based approaches and their applications to atherosclerosis plaque imaging and risk stratification. Finally, the challenges and opportunities regarding the future development and clinical translation of the targeted nanomedicine in related fields are discussed.
Collapse
Affiliation(s)
| | | | | | | | | | - Xuening Zhang
- Department of Radiology, Tianjin Medical University Second Hospital, Tianjin, China
| |
Collapse
|
21
|
Park J, Young BD, Miller EJ. Potential novel imaging targets of inflammation in cardiac sarcoidosis. J Nucl Cardiol 2022; 29:2171-2187. [PMID: 34734365 DOI: 10.1007/s12350-021-02838-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 09/26/2021] [Indexed: 10/19/2022]
Abstract
Cardiac sarcoidosis (CS) is an inflammatory disease with high morbidity and mortality, with a pathognomonic feature of non-caseating granulomatous inflammation. While 18F-fluorodeoxyglucose (FDG) positron emission tomography (PET) is a well-established modality to image inflammation and diagnose CS, there are limitations to its specificity and reproducibility. Imaging focused on the molecular processes of inflammation including the receptors and cellular microenvironments present in sarcoid granulomas provides opportunities to improve upon FDG-PET imaging for CS. This review will highlight the current limitations of FDG-PET imaging for CS while discussing emerging new nuclear imaging molecular targets for the imaging of cardiac sarcoidosis.
Collapse
Affiliation(s)
- Jakob Park
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Bryan D Young
- Section of Cardiovascular Medicine, Department of Medicine, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Edward J Miller
- Section of Cardiovascular Medicine, Department of Medicine, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA.
| |
Collapse
|
22
|
Lambertini A, Hartrampf PE, Higuchi T, Serfling SE, Meybohm P, Schirbel A, Buck AK, Werner RA. CXCR4-targeted molecular imaging after severe SARS-Cov-2 infection. Eur J Nucl Med Mol Imaging 2022; 50:228-229. [PMID: 35953728 PMCID: PMC9371958 DOI: 10.1007/s00259-022-05932-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 08/03/2022] [Indexed: 12/04/2022]
Affiliation(s)
- Alessandro Lambertini
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Philipp E Hartrampf
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Takahiro Higuchi
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, 97080, Würzburg, Germany.,Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Sebastian E Serfling
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Patrick Meybohm
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Wuerzburg, Würzburg, Germany
| | - Andreas Schirbel
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, 97080, Würzburg, Germany.
| | - Rudolf A Werner
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, 97080, Würzburg, Germany. .,Division of Nuclear Medicine and Molecular Imaging, The Russell H Morgan Department of Radiology and Radiological Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
23
|
Jiang CY, Zhong TT, Qiu LW, Liu YF, Zuo HH, Huang XF. The potential role of circulating exosomes in protecting myocardial injury in acute myocardial infarction via regulating miR-190a-3p/CXCR4/CXCL12 pathway. J Bioenerg Biomembr 2022; 54:175-189. [PMID: 35867293 DOI: 10.1007/s10863-022-09944-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 07/04/2022] [Indexed: 11/30/2022]
Abstract
Exosomes of different origins have been found to be protective against ischemic-induced myocardial injury. This study examined the protective effects of circulating exosomes in the mice model of acute myocardial infarction (AMI) and explored the underlying molecular mechanisms. The effects of exosomes on myocardial injury were assessed in the AMI mice model. The in vivo studies showed that circulating exosomes reduced the infarcted size, improved the morphology of heart tissues and also reduced apoptosis of the heart tissues. In addition, the model mice showed an increase in the CD34 + /VEGFR2 + cell population and CD31, CXCR4 and CXCL12 expression after exosomes treatment. MiR-190a-3p was significantly down-regulated in the exosomes derived from the culture medium of hypoxia-treated human cardiomyocytes (HCMs). Further analysis revealed that miR-190a-3p could physically interact with CXCR4/CXCL12 by targeting the respective 3'UTRs. These exosomes could up-regulated CXCR4 and CXCL12 expression in the EPCs; in addition, miR-190a-3p mimics repressed CXCR4/CXCL12 expression in EPCs, while its inhibitor had opposite effects. The in vitro functional assays showed that miR-190a-3p overexpression suppressed the cell viability, proliferation, migration, adhesion and tube formation of EPCs; while miR-190a-3p inhibitor had the opposite effects; exosomes derived from the culture medium of hypoxia-treated HCMs exhibited similar actions of miR-190a-3p inhibitor. Moreover, miR-190a-3p was down-regulated in exosomes from serum in the AMI group when compared to that from sham group. Treatment with exosomes from serum in the AMI group promoted cell proliferation, migration, adhesion and tube formation of EPCs when compared to that in the sham group. More importantly, IT1t attenuated the enhanced effects of miR-190a-3p inhibition on EPC proliferation, migration, adhesion and tube formation. In conclusion, circulating exosomes exerted protective effects on myocardial injury in the AMI mice model, and down-regulation of miR-190a-3p in the circulating exosomes may exert protective effects against myocardial injury. Hypoxia induced the downregulation of miR-190a-3p in the culture medium of HCMs, and the mechanistic investigations indicated that exosomes of hypoxia-conditioned HCM culture medium promoted the cell viability, proliferation, migration, adhesion and tube formation of EPCs via regulating miR-190a-3p/CXCR4/CXCL12 pathway.
Collapse
Affiliation(s)
- Chun-Yuan Jiang
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Jiangxi, 330006, Nanchang, China
| | - Ting-Ting Zhong
- Department of Cardiology, Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzhen, (Shenzhen Sun Yat-Sen Cardiovascular Hospital), Shenzhen, 518057, China.
| | - Lu-Wen Qiu
- Department of Cardiology, Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzhen, (Shenzhen Sun Yat-Sen Cardiovascular Hospital), Shenzhen, 518057, China
| | - Yan-Feng Liu
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Jiangxi, 330006, Nanchang, China
| | - Hui-Hua Zuo
- Department of Cardiology, Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzhen, (Shenzhen Sun Yat-Sen Cardiovascular Hospital), Shenzhen, 518057, China
| | - Xiao-Fei Huang
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Jiangxi, 330006, Nanchang, China
| |
Collapse
|
24
|
Stendahl JC, Kwan JM, Pucar D, Sadeghi MM. Radiotracers to Address Unmet Clinical Needs in Cardiovascular Imaging, Part 2: Inflammation, Fibrosis, Thrombosis, Calcification, and Amyloidosis Imaging. J Nucl Med 2022; 63:986-994. [PMID: 35772956 PMCID: PMC9258561 DOI: 10.2967/jnumed.121.263507] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 02/22/2022] [Indexed: 01/03/2023] Open
Abstract
Cardiovascular imaging is evolving in response to systemwide trends toward molecular characterization and personalized therapies. The development of new radiotracers for PET and SPECT imaging is central to addressing the numerous unmet diagnostic needs that relate to these changes. In this 2-part review, we discuss select radiotracers that may help address key unmet clinical diagnostic needs in cardiovascular medicine. Part 1 examined key technical considerations pertaining to cardiovascular radiotracer development and reviewed emerging radiotracers for perfusion and neuronal imaging. Part 2 covers radiotracers for imaging cardiovascular inflammation, thrombosis, fibrosis, calcification, and amyloidosis. These radiotracers have the potential to address several unmet needs related to the risk stratification of atheroma, detection of thrombi, and the diagnosis, characterization, and risk stratification of cardiomyopathies. In the first section, we discuss radiotracers targeting various aspects of inflammatory responses in pathologies such as myocardial infarction, myocarditis, sarcoidosis, atherosclerosis, and vasculitis. In a subsequent section, we discuss radiotracers for the detection of systemic and device-related thrombi, such as those targeting fibrin (e.g., 64Cu-labeled fibrin-binding probe 8). We also cover emerging radiotracers for the imaging of cardiovascular fibrosis, such as those targeting fibroblast activation protein (e.g., 68Ga-fibroblast activation protein inhibitor). Lastly, we briefly review radiotracers for imaging of cardiovascular calcification (18F-NaF) and amyloidosis (e.g., 99mTc-pyrophosphate and 18F-florbetapir).
Collapse
Affiliation(s)
- John C Stendahl
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Jennifer M Kwan
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Darko Pucar
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut; and
| | - Mehran M Sadeghi
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, Connecticut;
- Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| |
Collapse
|
25
|
Frantz S, Hundertmark MJ, Schulz-Menger J, Bengel FM, Bauersachs J. Left ventricular remodelling post-myocardial infarction: pathophysiology, imaging, and novel therapies. Eur Heart J 2022; 43:2549-2561. [PMID: 35511857 PMCID: PMC9336586 DOI: 10.1093/eurheartj/ehac223] [Citation(s) in RCA: 188] [Impact Index Per Article: 94.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 03/08/2022] [Accepted: 04/13/2022] [Indexed: 12/11/2022] Open
Abstract
Most patients survive acute myocardial infarction (MI). Yet this encouraging development has certain drawbacks: heart failure (HF) prevalence is increasing and patients affected tend to have more comorbidities worsening economic strain on healthcare systems and impeding effective medical management. The heart’s pathological changes in structure and/or function, termed myocardial remodelling, significantly impact on patient outcomes. Risk factors like diabetes, chronic obstructive pulmonary disease, female sex, and others distinctly shape disease progression on the ‘road to HF’. Despite the availability of HF drugs that interact with general pathways involved in myocardial remodelling, targeted drugs remain absent, and patient risk stratification is poor. Hence, in this review, we highlight the pathophysiological basis, current diagnostic methods and available treatments for cardiac remodelling following MI. We further aim to provide a roadmap for developing improved risk stratification and novel medical and interventional therapies.
Collapse
Affiliation(s)
- Stefan Frantz
- Department of Internal Medicine I, Universitätsklinikum Würzburg, University Hospital Würzburg, University of Würzburg, Oberdürrbacher Str. 6, 97080 Würzburg, Germany
| | - Moritz Jens Hundertmark
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Jeanette Schulz-Menger
- Department of Cardiology and Nephrology, Experimental and Clinical Research Center, a Joint Cooperation between the Charité Medical Faculty and the Max-Delbrueck Center for Molecular Medicine and HELIOS Hospital Berlin Buch, Berlin, Germany
| | | | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
26
|
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
|
27
|
Varasteh Z, Braeuer M, Mohanta S, Steinsiek AL, Habenicht A, Omidvari N, Topping GJ, Rischpler C, Weber WA, Sager HB, Raes G, Hernot S, Schwaiger M. In vivo Visualization of M2 Macrophages in the Myocardium After Myocardial Infarction (MI) Using 68Ga-NOTA-Anti-MMR Nb: Targeting Mannose Receptor (MR, CD206) on M2 Macrophages. Front Cardiovasc Med 2022; 9:889963. [PMID: 35548425 PMCID: PMC9081970 DOI: 10.3389/fcvm.2022.889963] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 03/29/2022] [Indexed: 12/02/2022] Open
Abstract
Introduction and Objectives Wound healing after myocardial infarction (MI) is a dynamic and complex multiple phase process, and a coordinated cellular response is required for proper scar formation. The current paradigm suggests that pro-inflammatory monocytes infiltrate the MI zone during the initial pro-inflammatory phase and differentiate into inflammatory macrophages, and then switch their phenotypes to anti-inflammatory during the reparative phase. Visualization of the reparative phase post-MI is of great interest because it may reveal delayed resolution of inflammation, which in turn predicts adverse cardiac remodeling. Imaging of anti-inflammatory macrophages may also be used to assess therapy approaches aiming to modulate the inflammatory response in order to limit MI size. Reparative macrophages can be distinguished from inflammatory macrophages by the surface marker mannose receptor (MR, CD206). In this study we evaluated the feasibility of 68Ga-NOTA-anti-MMR Nb for imaging of MR on alternatively activated macrophages in murine MI models. Methods Wildtype and MR-knockout mice and Wistar rats were subjected to MI via permanent ligation of the left coronary artery. Non-operated or sham-operated animals were used as controls. MR expression kinetics on cardiac macrophages was measured in mice using flow cytometry. PET/CT scans were performed 1 h after intravenous injection of 68Ga-NOTA-anti-MMR Nb. Mice and rats were euthanized and hearts harvested for ex vivo PET/MRI, autoradiography, and staining. As a non-targeting negative control, 68Ga-NOTA-BCII10 was used. Results In vivo-PET/CT scans showed focal radioactivity signals in the infarcted myocardium for 68Ga-NOTA-anti-MMR Nb which were confirmed by ex vivo-PET/MRI scans. In autoradiography images, augmented uptake of the tracer was observed in infarcts, as verified by the histochemistry analysis. Immunofluorescence staining demonstrated the presence and co-localization of CD206- and CD68-positive cells, in accordance to infarct zone. No in vivo or ex vivo signal was observed in the animals injected with control Nb or in the sham-operated animals. 68Ga-NOTA-anti-MMR Nb uptake in the infarcts of MR-knockout mice was negligibly low, confirming the specificity of 68Ga-NOTA-anti-MMR Nb to MR. Conclusion This exploratory study highlights the potential of 68Ga-NOTA-anti-MMR Nb to image MR-positive macrophages that are known to play a pivotal role in wound healing that follows acute MI.
Collapse
Affiliation(s)
- Zohreh Varasteh
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- *Correspondence: Zohreh Varasteh,
| | - Miriam Braeuer
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Sarajo Mohanta
- Institute for Cardiovascular Prevention, University Hospital of Ludwig-Maximilians-University, Munich, Germany
| | - Anna-Lena Steinsiek
- Department of Cardiology, German Heart Centre Munich, Technical of University Munich, Munich, Germany
| | - Andreas Habenicht
- Institute for Cardiovascular Prevention, University Hospital of Ludwig-Maximilians-University, Munich, Germany
| | - Negar Omidvari
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Geoffrey J. Topping
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Christoph Rischpler
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Wolfgang A. Weber
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Hendrik B. Sager
- Department of Cardiology, German Heart Centre Munich, Technical of University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Geert Raes
- Laboratory of Cellular and Molecular Immunology (CMIM), Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Sophie Hernot
- Laboratory for in vivo Cellular and Molecular Imaging, ICMI-BEFY/MIMA, Vrije Universiteit Brussel, Brussels, Belgium
| | - Markus Schwaiger
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| |
Collapse
|
28
|
Rurik JG, Epstein JA. Uniting Disciplines to Develop Therapeutics: Targeted mRNA Lipid Nanoparticles Reprogram the Immune System In Vivo to Treat Heart Disease. DNA Cell Biol 2022; 41:539-543. [PMID: 35446147 DOI: 10.1089/dna.2022.0171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The burgeoning field of immunomedicine is primed to expand beyond oncology (Aghajanian et al., 2022). Over the past several decades, many cell-based therapies have been proposed, developed, and deployed in the clinic. The recent explosion of targeted cell therapies has primarily been aimed at oncological malignancies. In parallel, cardiology researchers have been investigating the various cell types that contribute to heart diseases, especially those responsible for tissue fibrosis and myocardial dysfunction. Our laboratory proposed in 2019 to unite these two disciplines: could a targeted cell therapy be used to ameliorate cardiac fibrosis (Aghajanian et al., 2019). Although preliminary results were encouraging, the genetic engineering approach used to manufacture immune cells would result in persistent cytolytic T cell if directly translated to humans. This would pose a safety concern since activated fibroblasts are essential cells in the setting of acute injury. Therefore, we developed a novel technology to deliver modified RNA to T cells in vivo, resulting in a transient antiactivated fibroblast therapeutic (Rurik et al., 2022). Although active for only a few days, these cells were sufficient to significantly improve cardiac function in a murine model of cardiac fibrosis. These results pave the way for low-cost and scalable, and dose-able and immune therapy for fibrotic disorders.
Collapse
Affiliation(s)
- Joel G Rurik
- Department of Cell and Developmental Biology, Penn Cardiovascular Institute, and Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jonathan A Epstein
- Department of Cell and Developmental Biology, Penn Cardiovascular Institute, and Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
29
|
Stendahl JC, Liu Z, Boutagy NE, Nataneli E, Daghighian F, Sinusas AJ. Prototype device for endoventricular beta-emitting radiotracer detection and molecularly-guided intervention. J Nucl Cardiol 2022; 29:663-676. [PMID: 32820423 PMCID: PMC7895860 DOI: 10.1007/s12350-020-02317-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 07/10/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND We have set out to develop a catheter-based theranostic system that: (a) identifies diseased and at-risk myocardium via endocardial detection of systemically delivered β-emitting radiotracers and (b) utilizes molecular signals to guide delivery of therapeutics to appropriate tissue via direct intramyocardial injection. METHODS Our prototype device consists of a miniature β-radiation detector contained within the tip of a flexible intravascular catheter. The catheter can be adapted to incorporate an injection port and retractable needle for therapeutic delivery. The performance of the β-detection catheter was assessed in vitro with various β-emitting radionuclides and ex vivo in hearts of pigs following systemic injection of 18F-fluorodeoxyglucose (18F-FDG) at 1-week post-myocardial infarction. Regional catheter-based endocardial measurements of 18F activity were compared to regional tissue activity from PET/CT images and gamma counting. RESULTS The β-detection catheter demonstrated sensitive in vitro detection of β-radiation from 22Na (β+), 18F (β+), and 204Tl (β-), with minimal sensitivity to γ-radiation. For 18F, the catheter demonstrated a sensitivity of 4067 counts/s/μCi in contact and a spatial resolution of 1.1 mm FWHM. Ex vivo measurements of endocardial 18F activity with the β-detection catheter in the chronic pig infarct model demonstrated good qualitative and quantitative correlation with regional tissue activity from PET/CT images and gamma counting. CONCLUSION The prototype β-detection catheter demonstrates sensitive and selective detection of β- and β+ emissions over a wide range of energies and enables high-fidelity ex vivo characterization of endocardial activity from systemically delivered 18F-FDG.
Collapse
Affiliation(s)
- John C Stendahl
- Section of Cardiovascular Medicine, Department of Medicine, Yale Translational Research Imaging Center, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Zhao Liu
- Department of Biomedical Engineering, Yale University, School of Engineering and Applied Science, New Haven, CT, 06520, USA
| | - Nabil E Boutagy
- Section of Cardiovascular Medicine, Department of Medicine, Yale Translational Research Imaging Center, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Eliahoo Nataneli
- IntraMedical Imaging, LLC, 12569 Crenshaw Blvd, Hawthorne, CA, 90250, USA
| | - Farhad Daghighian
- IntraMedical Imaging, LLC, 12569 Crenshaw Blvd, Hawthorne, CA, 90250, USA
| | - Albert J Sinusas
- Section of Cardiovascular Medicine, Department of Medicine, Yale Translational Research Imaging Center, Yale University School of Medicine, New Haven, CT, 06520, USA.
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, P.O. Box 208017, Dana 3, New Haven, CT, 06520-8017, USA.
- Department of Biomedical Engineering, Yale University, School of Engineering and Applied Science, New Haven, CT, 06520, USA.
| |
Collapse
|
30
|
Schäfer A, Akin M, Diekmann J, König T. Intracoronary Application of Super-Saturated Oxygen to Reduce Infarct Size Following Myocardial Infarction. J Clin Med 2022; 11:jcm11061509. [PMID: 35329835 PMCID: PMC8949147 DOI: 10.3390/jcm11061509] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/03/2022] [Accepted: 03/08/2022] [Indexed: 01/27/2023] Open
Abstract
Optimal medical therapy for secondary prevention following acute myocardial infarction reduces non-fatal ischaemic events. Intensive antithrombotic or lipid-lowering approaches have failed to significantly lower mortality. In the past, reduction of infarct size in patients undergoing primary percutaneous revascularisation for acute myocardial infarction had been considered as a surrogate outcome marker. However, infarct size measured by magnetic resonance imaging or SPECT is strongly associated with all-cause mortality and hospitalization for heart failure within the first year after an acute myocardial infarction. Intracoronary administration of super-saturated oxygen (SSO2) immediately after revascularisation is an approach that can be used to reduce infarct size and, therefore, improve cardiovascular outcome in patients with acute myocardial infarction. In this article, we describe the modulation of pathophysiology by SSO2, review the existing trial data and present our first impressions with the technique in real clinical practice.
Collapse
Affiliation(s)
- Andreas Schäfer
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; (M.A.); (T.K.)
- Correspondence: ; Tel.: +49-(511)-532-5240
| | - Muharrem Akin
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; (M.A.); (T.K.)
| | - Johanna Diekmann
- Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany;
| | - Tobias König
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; (M.A.); (T.K.)
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW Successful treatment of cancer can be hampered by the attendant risk of cardiotoxicity, manifesting as cardiomyopathy, left ventricle systolic dysfunction and, in some cases, heart failure. This risk can be mitigated if the injury to the heart is detected before the onset to irreversible cardiac impairment. The gold standard for cardiac imaging in cardio-oncology is echocardiography. Despite improvements in the application of this modality, it is not typically sensitive to sub-clinical or early-stage dysfunction. We identify in this review some emerging tracers for detecting incipient cardiotoxicity by positron emission tomography (PET). RECENT FINDINGS Vectors labeled with positron-emitting radionuclides (e.g., carbon-11, fluorine-18, gallium-68) are now available to study cardiac function, metabolism, and tissue repair in preclinical models. Many of these probes are highly sensitive to early damage, thereby potentially addressing the limitations of current imaging approaches, and show promise in preliminary clinical evaluations. The overlapping pathophysiology between cardiotoxicity and heart failure significantly expands the number of imaging tools available to cardio-oncology. This is highlighted by the emergence of radiolabeled probes targeting fibroblast activation protein (FAP) for sensitive detection of dysregulated healing process that underpins adverse cardiac remodeling. The growth of PET scanner technology also creates an opportunity for a renaissance in metabolic imaging in cardio-oncology research.
Collapse
Affiliation(s)
- James M. Kelly
- Division of Radiopharmaceutical Sciences and Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, Belfer Research Building, Room BB-1604, 413 East 69th St, New York, NY 10021 USA
- Citigroup Biomedical Imaging Center, Weill Cornell Medicine, New York, NY 10021 USA
| | - John W. Babich
- Division of Radiopharmaceutical Sciences and Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, Belfer Research Building, Room BB-1604, 413 East 69th St, New York, NY 10021 USA
- Citigroup Biomedical Imaging Center, Weill Cornell Medicine, New York, NY 10021 USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021 USA
| |
Collapse
|
32
|
Rogasch JMM, Hofheinz F, van Heek L, Voltin CA, Boellaard R, Kobe C. Influences on PET Quantification and Interpretation. Diagnostics (Basel) 2022; 12:451. [PMID: 35204542 PMCID: PMC8871060 DOI: 10.3390/diagnostics12020451] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/06/2022] [Accepted: 02/08/2022] [Indexed: 01/21/2023] Open
Abstract
Various factors have been identified that influence quantitative accuracy and image interpretation in positron emission tomography (PET). Through the continuous introduction of new PET technology-both imaging hardware and reconstruction software-into clinical care, we now find ourselves in a transition period in which traditional and new technologies coexist. The effects on the clinical value of PET imaging and its interpretation in routine clinical practice require careful reevaluation. In this review, we provide a comprehensive summary of important factors influencing quantification and interpretation with a focus on recent developments in PET technology. Finally, we discuss the relationship between quantitative accuracy and subjective image interpretation.
Collapse
Affiliation(s)
- Julian M. M. Rogasch
- Department of Nuclear Medicine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany;
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, 10178 Berlin, Germany
| | - Frank Hofheinz
- Institute of Radiopharmaceutical Cancer Research, Helmholtz Center Dresden-Rossendorf, 01328 Dresden, Germany;
| | - Lutz van Heek
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (L.v.H.); (C.-A.V.)
| | - Conrad-Amadeus Voltin
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (L.v.H.); (C.-A.V.)
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam (CCA), Amsterdam University Medical Center, Free University Amsterdam, 1081 HV Amsterdam, The Netherlands;
| | - Carsten Kobe
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (L.v.H.); (C.-A.V.)
| |
Collapse
|
33
|
Nuclear Molecular Imaging of Cardiac Remodeling after Myocardial Infarction. Pharmaceuticals (Basel) 2022; 15:ph15020183. [PMID: 35215296 PMCID: PMC8875369 DOI: 10.3390/ph15020183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 01/28/2022] [Accepted: 01/28/2022] [Indexed: 12/03/2022] Open
Abstract
The role of molecular imaging technologies in detecting, evaluating, and monitoring cardiovascular disease and their treatment is expanding rapidly. Gradually replacing the conventional anatomical or physiological approaches, molecular imaging strategies using biologically targeted markers provide unique insight into pathobiological processes at molecular and cellular levels and allow for cardiovascular disease evaluation and individualized therapy. This review paper will discuss currently available and developing molecular-based single-photon emission computed tomography (SPECT) and positron emission tomography (PET) imaging strategies to evaluate post-infarction cardiac remodeling. These approaches include potential targeted methods of evaluating critical biological processes, such as inflammation, angiogenesis, and scar formation.
Collapse
|
34
|
Zhu Z, Zhang G, Li D, Yin X, Wang T. Silencing of specificity protein 1 protects H9c2 cells against lipopolysaccharide-induced injury via binding to the promoter of chemokine CXC receptor 4 and suppressing NF-κB signaling. Bioengineered 2022; 13:3395-3409. [PMID: 35048778 PMCID: PMC8973921 DOI: 10.1080/21655979.2022.2026548] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
G protein-coupled protein receptor CXC chemokine receptor 4 (CXCR4) has been shown to be involved in the development of sepsis; however, it remains unclear whether CXCR4 participates in the septic myocardial injury. In our study, treatment with lipopolysaccharide (LPS) increased the expression of specificity protein 1 (SP1) and CXCR4 in H9c2 cells. Notably, a positive association between SP1 and CXCR4 expression was observed in LPS-treated H9c2 cells, and SP1 positively regulated CXCR4 expression in H9c2 cells. Moreover, silencing of SP1 or CXCR4 suppressed LPS-induced inflammation and cell apoptosis in H9c2 cells, as evidenced by the increase in cell viability and decrease in lactate dehydrogenase release, interleukin (IL)-6, IL-8, and tumor necrosis factor (TNF)-α levels, and caspase-3 activity. Additionally, overexpression of CXCR4 abolished the protective effects of SP1 silencing on LPS-induced injury in H9c2 cells. SP1 was also shown to enhance the promoter activity of CXCR4 by directly binding with the binding motif site – 109/–100 in CXCR4 promoter. Besides, downregulation of SP1 or CXCR4 blocked LPS-induced activation of the NF-кB signaling in H9c2 cells. Furthermore, inhibition of NF-кB signaling by DHMEQ abolished LPS-induced myocardial inflammation and apoptosis. In conclusion, silencing of SP1 protected H9c2 cells against LPS-induced injury by binding to the promoter of CXCR4 and suppressing the NF-κB signaling pathway. Hence, our findings provide evidence that manipulation of SP1 or CXCR4 may be an effective approach to promote prevention or recovery of septic myocardial injury, and thereby, may serve as a potential therapeutic strategy for sepsis.
Collapse
Affiliation(s)
- Zhao Zhu
- Department of Emergency, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003 China
| | - Guoxiu Zhang
- Department of Emergency, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003 China
| | - Dahuan Li
- Department of Emergency, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003 China
| | - Xiaojun Yin
- Department of Emergency, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003 China
| | - Tianzhong Wang
- Department of Emergency, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003 China
| |
Collapse
|
35
|
Atypical Roles of the Chemokine Receptor ACKR3/CXCR7 in Platelet Pathophysiology. Cells 2022; 11:cells11020213. [PMID: 35053329 PMCID: PMC8773869 DOI: 10.3390/cells11020213] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 12/23/2022] Open
Abstract
The manifold actions of the pro-inflammatory and regenerative chemokine CXCL12/SDF-1α are executed through the canonical GProteinCoupledReceptor CXCR4, and the non-canonical ACKR3/CXCR7. Platelets express CXCR4, ACKR3/CXCR7, and are a vital source of CXCL12/SDF-1α themselves. In recent years, a regulatory impact of the CXCL12-CXCR4-CXCR7 axis on platelet biogenesis, i.e., megakaryopoiesis, thrombotic and thrombo-inflammatory actions have been revealed through experimental and clinical studies. Platelet surface expression of ACKR3/CXCR7 is significantly enhanced following myocardial infarction (MI) in acute coronary syndrome (ACS) patients, and is also associated with improved functional recovery and prognosis. The therapeutic implications of ACKR3/CXCR7 in myocardial regeneration and improved recovery following an ischemic episode, are well documented. Cardiomyocytes, cardiac-fibroblasts, endothelial lining of the blood vessels perfusing the heart, besides infiltrating platelets and monocytes, all express ACKR3/CXCR7. This review recapitulates ligand induced differential trafficking of platelet CXCR4-ACKR3/CXCR7 affecting their surface availability, and in regulating thrombo-inflammatory platelet functions and survival through CXCR4 or ACKR3/CXCR7. It emphasizes the pro-thrombotic influence of CXCL12/SDF-1α exerted through CXCR4, as opposed to the anti-thrombotic impact of ACKR3/CXCR7. Offering an innovative translational perspective, this review also discusses the advantages and challenges of utilizing ACKR3/CXCR7 as a potential anti-thrombotic strategy in platelet-associated cardiovascular disorders, particularly in coronary artery disease (CAD) patients post-MI.
Collapse
|
36
|
Molecular imaging of the brain-heart axis provides insights into cardiac dysfunction after cerebral ischemia. Basic Res Cardiol 2022; 117:52. [PMID: 36279013 PMCID: PMC9592646 DOI: 10.1007/s00395-022-00961-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/26/2022] [Accepted: 10/13/2022] [Indexed: 01/31/2023]
Abstract
Ischemic stroke imparts elevated risk of heart failure though the underlying mechanisms remain poorly described. We aimed to characterize the influence of cerebral ischemic injury on cardiac function using multimodality molecular imaging to investigate brain and cardiac morphology and tissue inflammation in two mouse models of variable stroke severity. Transient middle cerebral artery occlusion (MCAo) generated extensive stroke damage (56.31 ± 40.39 mm3). Positron emission tomography imaging of inflammation targeting the mitochondrial translocator protein (TSPO) revealed localized neuroinflammation at 7 days after stroke compared to sham (3.8 ± 0.8 vs 2.6 ± 0.7 %ID/g max, p < 0.001). By contrast, parenchyma topical application of vasoconstrictor endothelin-1 did not generate significant stroke damage or neuroinflammatory cell activity. MCAo evoked a modest reduction in left ventricle ejection fraction at both 1 weeks and 3 weeks after stroke (LVEF at 3 weeks: 54.3 ± 5.7 vs 66.1 ± 3.5%, p < 0.001). This contractile impairment was paralleled by elevated cardiac TSPO PET signal compared to sham (8.6 ± 2.4 vs 5.8 ± 0.7%ID/g, p = 0.022), but was independent of leukocyte infiltration defined by flow cytometry. Stroke size correlated with severity of cardiac dysfunction (r = 0.590, p = 0.008). Statistical parametric mapping identified a direct association between neuroinflammation at 7 days in a cluster of voxels including the insular cortex and reduced ejection fraction (ρ = - 0.396, p = 0.027). Suppression of microglia led to lower TSPO signal at 7 days which correlated with spared late cardiac function after MCAo (r = - 0.759, p = 0.029). Regional neuroinflammation early after cerebral ischemia influences subsequent cardiac dysfunction. Total body TSPO PET enables monitoring of neuroinflammation, providing insights into brain-heart inter-organ communication and may guide therapeutic intervention to spare cardiac function post-stroke.
Collapse
|
37
|
Hess A, Borchert T, Ross TL, Bengel FM, Thackeray JT. Characterizing the transition from immune response to tissue repair after myocardial infarction by multiparametric imaging. Basic Res Cardiol 2022; 117:14. [PMID: 35275268 PMCID: PMC8917105 DOI: 10.1007/s00395-022-00922-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/01/2022] [Accepted: 02/16/2022] [Indexed: 01/31/2023]
Abstract
Persistent inflammation following myocardial infarction (MI) precipitates adverse outcome including acute ventricular rupture and chronic heart failure. Molecular imaging allows longitudinal assessment of immune cell activity in the infarct territory and predicts severity of remodeling. We utilized a multiparametric imaging platform to assess the immune response and cardiac healing following MI in mice. Suppression of circulating macrophages prior to MI paradoxically resulted in higher total leukocyte content in the heart, demonstrated by increased CXC motif chemokine receptor 4 (CXCR4) positron emission tomography imaging. This supported the formation of a thrombus overlying the injured region, as identified by magnetic resonance imaging. The injured and thrombotic region in macrophage depeleted mice subsequently showed active calcification, as evidenced by accumulation of 18F-fluoride and by cardiac computed tomography. Importantly, macrophage suppression triggered a prolonged inflammatory response confirmed by post-mortem tissue analysis that was associated with higher mortality from ventricular rupture early after occlusion and with increased infarct size and worse chronic contractile function at 6 weeks after reperfusion. These findings establish a molecular imaging toolbox for monitoring the interplay between adverse immune response and tissue repair after MI. This may serve as a foundation for development and monitoring of novel targeted therapies that may include immune modulation and endogenous healing support.
Collapse
Affiliation(s)
- Annika Hess
- grid.10423.340000 0000 9529 9877Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Tobias Borchert
- grid.10423.340000 0000 9529 9877Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany ,Present Address: Cardior Pharmaceuticals GmbH, Hannover, Germany
| | - Tobias L. Ross
- grid.10423.340000 0000 9529 9877Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Frank M. Bengel
- grid.10423.340000 0000 9529 9877Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - James T. Thackeray
- grid.10423.340000 0000 9529 9877Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| |
Collapse
|
38
|
OUP accepted manuscript. Eur Heart J Cardiovasc Imaging 2022; 23:465-475. [DOI: 10.1093/ehjci/jeab287] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 12/21/2021] [Indexed: 11/13/2022] Open
|
39
|
Kalia N. A historical review of experimental imaging of the beating heart coronary microcirculation in vivo. J Anat 2021; 242:3-16. [PMID: 34905637 PMCID: PMC9773169 DOI: 10.1111/joa.13611] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/25/2021] [Accepted: 12/03/2021] [Indexed: 12/25/2022] Open
Abstract
Following a myocardial infarction (MI), the prognosis of patients is highly dependent upon the re-establishment of perfusion not only in the occluded coronary artery, but also within the coronary microcirculation. However, our fundamental understanding of the pathophysiology of the tiniest blood vessels of the heart is limited primarily because no current clinical imaging tools can directly visualise them. Moreover, in vivo experimental studies of the beating heart using intravital imaging have also been hampered due to obvious difficulties related to significant inherent contractile motion, movement of the heart brought about by nearby lungs and its location in an anatomically challenging position for microscopy. However, recent advances in microscopy techniques, and the development of fluorescent reporter mice and fluorescently conjugated antibodies allowing visualisation of vascular structures, thromboinflammatory cells and blood flow, have allowed us to overcome some of these challenges and increase our basic understanding of cardiac microvascular pathophysiology. In this review, the elegant attempts of the pioneers in intravital imaging of the beating heart will be discussed, which focussed on providing new insights into the anatomy and physiology of the healthy heart microvessels. The reviews end with the more recent studies that focussed on disease pathology and increasing our understanding of myocardial thromboinflammatory cell recruitment and flow disturbances, particularly in the setting of diseases such as MI.
Collapse
Affiliation(s)
- Neena Kalia
- Microcirculation Research GroupInstitute of Cardiovascular SciencesCollege of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| |
Collapse
|
40
|
PET Imaging in Cardiac Sarcoidosis: A Narrative Review with Focus on Novel PET Tracers. Pharmaceuticals (Basel) 2021; 14:ph14121286. [PMID: 34959686 PMCID: PMC8704408 DOI: 10.3390/ph14121286] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 01/04/2023] Open
Abstract
Sarcoidosis is a multi-system inflammatory disease characterized by the development of inflammation and noncaseating granulomas that can involve nearly every organ system, with a predilection for the pulmonary system. Cardiac involvement of sarcoidosis (CS) occurs in up to 70% of cases, and accounts for a significant share of sarcoid-related mortality. The clinical presentation of CS can range from absence of symptoms to conduction abnormalities, heart failure, arrhythmias, valvular disease, and sudden cardiac death. Given the significant morbidity and mortality associated with CS, timely diagnosis is important. Traditional imaging modalities and histologic evaluation by endomyocardial biopsy often provide a low diagnostic yield. Cardiac positron emission tomography (PET) has emerged as a leading advanced imaging modality for the diagnosis and management of CS. This review article will summarize several aspects of the current use of PET in CS, including indications for use, patient preparation, image acquisition and interpretation, diagnostic and prognostic performance, and evaluation of treatment response. Additionally, this review will discuss novel PET radiotracers currently under study or of potential interest in CS.
Collapse
|
41
|
Zacherl MJ, Todica A, Wängler C, Schirrmacher R, Hajebrahimi MA, Pircher J, Li X, Lindner S, Brendel M, Bartenstein P, Massberg S, Brunner S, Lehner S, Hacker M, Huber BC. Molecular imaging of cardiac CXCR4 expression in a mouse model of acute myocardial infarction using a novel 68Ga-mCXCL12 PET tracer. J Nucl Cardiol 2021; 28:2965-2975. [PMID: 32676914 PMCID: PMC8709820 DOI: 10.1007/s12350-020-02262-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 06/08/2020] [Indexed: 01/16/2023]
Abstract
BACKGROUND The chemokine receptor CXCR4 and its ligand CXCL12 have been shown to be a possible imaging and therapeutic target after myocardial infarction (MI). The murine-based and mouse-specific 68Ga-mCXCL12 PET tracer could be suitable for serial in vivo quantification of cardiac CXCR4 expression in a murine model of MI. METHODS AND RESULTS At days 1-6 after MI, mice were intravenously injected with 68Ga-mCXCL12. Autoradiography was performed and the infarct-to-remote ratio (I/R) was determined. In vivo PET imaging with 68Ga-mCXCL12 was conducted on days 1-6 after MI and the percentage of the injected dose (%ID/g) of the tracer uptake in the infarct area was calculated. 18F-FDG-PET was performed for anatomical landmarking. Ex vivo autoradiography identified CXCR4 upregulation in the infarct region with an increasing I/R after 12 hours (1.4 ± 0.3), showing a significant increase until day 2 (4.5 ± 0.6), followed by a plateau phase (day 4) and decrease after 10 days (1.3 ± 1.0). In vivo PET imaging identified similar CXCR4 upregulation in the infarct region which peaked around day 3 post MI (9.7 ± 5.0 %ID/g) and then subsequently decreased by day 6 (2.8 ± 1.0 %ID/g). CONCLUSION Noninvasive molecular imaging of cardiac CXCR4 expression using a novel, murine-based, and specific 68Ga-mCXCL12 tracer is feasible both ex vivo and in vivo.
Collapse
Affiliation(s)
| | - Andrei Todica
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Carmen Wängler
- Biomedical Chemistry, Department of Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Mannheim, Germany
| | - Ralf Schirrmacher
- Department of Oncology, Division of Oncological Imaging, University of Alberta, Edmonton, AB, Canada
| | | | - Joachim Pircher
- Department of Cardiology, University Hospital of Munich, LMU Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Xiang Li
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Simon Lindner
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Steffen Massberg
- Department of Cardiology, University Hospital of Munich, LMU Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Stefan Brunner
- Department of Cardiology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Sebastian Lehner
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- Ambulatory Healthcare Center Dr. Neumaier & Colleagues, Radiology, Nuclear Medicine, Radiation Therapy, Regensburg, Germany
| | - Marcus Hacker
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria.
| | - Bruno C Huber
- Department of Cardiology, University Hospital of Munich, LMU Munich, Munich, Germany
| |
Collapse
|
42
|
Feher A, Sinusas AJ. Evaluation of cardiac allograft vasculopathy by positron emission tomography. J Nucl Cardiol 2021; 28:2616-2628. [PMID: 33389637 DOI: 10.1007/s12350-020-02438-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/04/2020] [Indexed: 12/22/2022]
Abstract
Cardiac allograft vasculopathy (CAV) remains one of the most important late occurring complications in heart transplant (HT) recipients significantly effecting graft survival. Recently, there has been tremendous focus on the development of effective and safe non-invasive diagnostic strategies for the diagnosis of CAV employing a wide range of imaging technologies. During the past decade multiple studies have been published using positron emission tomography (PET) myocardial perfusion imaging, establishing the value of PET myocardial blood flow quantification for the evaluation of CAV. These independent investigations demonstrate that PET can be successfully used to establish the diagnosis of CAV, can be utilized for prognostication and may be used for serial monitoring of HT recipients. In addition, molecular imaging techniques have started to emerge as new tools to enhance our knowledge to better understand the pathophysiology of CAV.
Collapse
Affiliation(s)
- Attila Feher
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, P.O. Box 208017, Dana 3, New Haven, CT, 06520, USA.
| | - Albert J Sinusas
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, P.O. Box 208017, Dana 3, New Haven, CT, 06520, USA
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| |
Collapse
|
43
|
Schottelius M, Herrmann K, Lapa C. In Vivo Targeting of CXCR4-New Horizons. Cancers (Basel) 2021; 13:5920. [PMID: 34885030 PMCID: PMC8656854 DOI: 10.3390/cancers13235920] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/22/2021] [Accepted: 11/22/2021] [Indexed: 01/23/2023] Open
Abstract
Given its pre-eminent role in the context of tumor cell growth as well as metastasis, the C-X-C motif chemokine receptor 4 (CXCR4) has attracted a lot of interest in the field of nuclear oncology, and clinical evidence on the high potential of CXCR4-targeted theranostics is constantly accumulating. Additionally, since CXCR4 also represents a key player in the orchestration of inflammatory responses to inflammatory stimuli, based on its expression on a variety of pro- and anti-inflammatory immune cells (e.g., macrophages and T-cells), CXCR4-targeted inflammation imaging has recently gained considerable attention. Therefore, after briefly summarizing the current clinical status quo of CXCR4-targeted theranostics in cancer, this review primarily focuses on imaging of a broad spectrum of inflammatory diseases via the quantification of tissue infiltration with CXCR4-expressing immune cells. An up-to-date overview of the ongoing preclinical and clinical efforts to visualize inflammation and its resolution over time is provided, and the predictive value of the CXCR4-associated imaging signal for disease outcome is discussed. Since the sensitivity and specificity of CXCR4-targeted immune cell imaging greatly relies on the availability of suitable, tailored imaging probes, recent developments in the field of CXCR4-targeted imaging agents for various applications are also addressed.
Collapse
Affiliation(s)
- Margret Schottelius
- Translational Radiopharmaceutical Sciences, Department of Nuclear Medicine and of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne (UNIL), 1011 Lausanne, Switzerland
| | - Ken Herrmann
- Department of Nuclear Medicine, German Cancer Consortium (DKTK)-University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany;
| | - Constantin Lapa
- Nuclear Medicine, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany
| |
Collapse
|
44
|
Prigent K, Vigne J. Advances in Radiopharmaceutical Sciences for Vascular Inflammation Imaging: Focus on Clinical Applications. Molecules 2021; 26:molecules26237111. [PMID: 34885690 PMCID: PMC8659223 DOI: 10.3390/molecules26237111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/09/2021] [Accepted: 11/19/2021] [Indexed: 01/18/2023] Open
Abstract
Biomedical imaging technologies offer identification of several anatomic and molecular features of disease pathogenesis. Molecular imaging techniques to assess cellular processes in vivo have been useful in advancing our understanding of several vascular inflammatory diseases. For the non-invasive molecular imaging of vascular inflammation, nuclear medicine constitutes one of the best imaging modalities, thanks to its high sensitivity for the detection of probes in tissues. 2-[18F]fluoro-2-deoxy-d-glucose ([18F]FDG) is currently the most widely used radiopharmaceutical for molecular imaging of vascular inflammatory diseases such as atherosclerosis and large-vessel vasculitis. The combination of [18F]FDG and positron emission tomography (PET) imaging has become a powerful tool to identify and monitor non-invasively inflammatory activities over time but suffers from several limitations including a lack of specificity and avid background in different localizations. The use of novel radiotracers may help to better understand the underlying pathophysiological processes and overcome some limitations of [18F]FDG PET for the imaging of vascular inflammation. This review examines how [18F]FDG PET has given us deeper insight into the role of inflammation in different vascular pathologies progression and discusses perspectives for alternative radiopharmaceuticals that could provide a more specific and simple identification of pathologies where vascular inflammation is implicated. 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. Future research is needed to realize the true clinical translational value of PET imaging in vascular inflammatory diseases.
Collapse
Affiliation(s)
- Kevin Prigent
- CHU de Caen Normandie, Department of Nuclear Medicine, Normandie Université, UNICAEN, 14000 Caen, France;
| | - Jonathan Vigne
- CHU de Caen Normandie, Department of Nuclear Medicine, Normandie Université, UNICAEN, 14000 Caen, France;
- CHU de Caen Normandie, Department of Pharmacy, Normandie Université, UNICAEN, 14000 Caen, France
- UNICAEN, INSERM U1237, Etablissement Français du Sang, Physiopathology and Imaging of Neurological Disorders (PhIND), Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Normandie University, 14000 Caen, France
- Correspondence:
| |
Collapse
|
45
|
CXCR4-Targeted Imaging of Post-Infarct Myocardial Tissue Inflammation: Prognostic Value After Reperfused Myocardial Infarction. JACC Cardiovasc Imaging 2021; 15:372-374. [PMID: 34656473 DOI: 10.1016/j.jcmg.2021.08.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/09/2021] [Accepted: 08/23/2021] [Indexed: 11/22/2022]
|
46
|
Yu X, Newland SA, Zhao TX, Lu Y, Sage AS, Sun Y, Sriranjan RS, Ma MKL, Lam BYH, Nus M, Harrison JE, Bond SJ, Cheng X, Silvestre JS, Rudd JHF, Cheriyan J, Mallat Z. Innate Lymphoid Cells Promote Recovery of Ventricular Function After Myocardial Infarction. J Am Coll Cardiol 2021; 78:1127-1142. [PMID: 34503682 PMCID: PMC8434674 DOI: 10.1016/j.jacc.2021.07.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 07/06/2021] [Accepted: 07/13/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND Innate lymphoid cells type 2 (ILC2s) play critical homeostatic functions in peripheral tissues. ILC2s reside in perivascular niches and limit atherosclerosis development. OBJECTIVES ILC2s also reside in the pericardium but their role in postischemic injury is unknown. METHODS We examined the role of ILC2 in a mouse model of myocardial infarction (MI), and compared mice with or without genetic deletion of ILC2. We determined infarct size using histology and heart function using echocardiography. We assessed cardiac ILC2 using flow cytometry and RNA sequencing. Based on these data, we devised a therapeutic strategy to activate ILC2 in mice with acute MI, using exogenous interleukin (IL)-2. We also assessed the ability of low-dose IL-2 to activate ILC2 in a double-blind randomized clinical trial of patients with acute coronary syndromes (ACS). RESULTS We found that ILC2 levels were increased in pericardial adipose tissue after experimental MI, and genetic ablation of ILC2 impeded the recovery of heart function. RNA sequencing revealed distinct transcript signatures in ILC2, and pointed to IL-2 axis as a major upstream regulator. Treatment of T-cell-deficient mice with IL-2 (to activate ILC2) significantly improved the recovery of heart function post-MI. Administration of low-dose IL-2 to patients with ACS led to activation of circulating ILC2, with significant increase in circulating IL-5, a prototypic ILC2-derived cytokine. CONCLUSIONS ILC2s promote cardiac healing and improve the recovery of heart function after MI in mice. Activation of ILC2 using low-dose IL-2 could be a novel therapeutic strategy to promote a reparative response after MI.
Collapse
Affiliation(s)
- Xian Yu
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom; Department of Cardiology, Union Hospital, Tongji, Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Stephen A Newland
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Tian X Zhao
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Yuning Lu
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Andrew S Sage
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Yanyi Sun
- Université de Paris, PARCC, INSERM, F-75015 Paris, France
| | - Rouchelle S Sriranjan
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Marcella K L Ma
- The Wellcome Trust-MRC Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, United Kingdom
| | - Brian Y H Lam
- The Wellcome Trust-MRC Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, United Kingdom
| | - Meritxell Nus
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - James E Harrison
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Simon J Bond
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, United Kingdom
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, Tongji, Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | - James H F Rudd
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Joseph Cheriyan
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, United Kingdom; Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Ziad Mallat
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom; Université de Paris, PARCC, INSERM, F-75015 Paris, France.
| |
Collapse
|
47
|
Du L, Chen J, Wu Y, Xia G, Chen M, Zhao P, Wang Y, Yao D, Liu F, Zhang L, Wang X, Yang Y, Wang L. Long Non-coding RNA N1LR Protects Against Myocardial Ischemic/Reperfusion Injury Through Regulating the TGF-β Signaling Pathway. Front Cardiovasc Med 2021; 8:654969. [PMID: 34485393 PMCID: PMC8414635 DOI: 10.3389/fcvm.2021.654969] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 07/12/2021] [Indexed: 11/13/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) have been shown to play critical roles in various cell biological processes. However, the mechanism of lncRNAs in acute myocardial infarction (AMI) is not fully understood. Previous studies showed that lncRNA N1LR was down-regulated in ischemic cerebral stroke and its up-regulation was protective. The current study was designed to assess the protective effect of N1LR and further to explore potential mechanisms of N1LR in ischemic/reperfusion (I/R) injury after AMI. Male C57BL/6J mice and H9c2 cardiomyocytes were selected to construct in vivo and in vitro pathological models. In H9c2 cell line, N1LR expression was markedly decreased after H2O2 and CoCl2 treatments and N1LR overexpression alleviated apoptosis, inflammation reaction, and LDH release in cardiomyocytes treated with H2O2 and CoCl2. Mouse in vivo study showed that overexpression of N1LR enhanced cardiac function and suppressed inflammatory response and fibrosis. Mechanistically, we found that the expression of transforming growth factor (TGF)-β1 and smads were significantly decreased in the N1LR overexpression group exposed to H2O2. In a summary, our study indicated that N1LR can act as a protective factor against cardiac ischemic-reperfusion injury through regulating the TGF-β/Smads signaling pathway.
Collapse
Affiliation(s)
- Lin Du
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China.,Department of Cardiology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Jie Chen
- Department of Gastroenterology, Northern Jiangsu Province People's Hospital, Yangzhou University, Yangzhou, China
| | - Yong Wu
- Department of Cardiology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Guangwei Xia
- Department of Cardiology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Mingxing Chen
- Department of Cardiology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Pei Zhao
- Department of Cardiology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Yao Wang
- Department of Cardiology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Deshan Yao
- Department of Cardiology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Fan Liu
- Department of Cardiology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Lina Zhang
- Department of Cardiology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Xue Wang
- Department of Cardiology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Yi Yang
- Department of Cardiology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Liansheng Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| |
Collapse
|
48
|
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
|
49
|
Alluri SR, Higashi Y, Kil KE. PET Imaging Radiotracers of Chemokine Receptors. Molecules 2021; 26:molecules26175174. [PMID: 34500609 PMCID: PMC8434599 DOI: 10.3390/molecules26175174] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/24/2021] [Accepted: 08/24/2021] [Indexed: 12/12/2022] Open
Abstract
Chemokines and chemokine receptors have been recognized as critical signal components that maintain the physiological functions of various cells, particularly the immune cells. The signals of chemokines/chemokine receptors guide various leukocytes to respond to inflammatory reactions and infectious agents. Many chemokine receptors play supportive roles in the differentiation, proliferation, angiogenesis, and metastasis of diverse tumor cells. In addition, the signaling functions of a few chemokine receptors are associated with cardiac, pulmonary, and brain disorders. Over the years, numerous promising molecules ranging from small molecules to short peptides and antibodies have been developed to study the role of chemokine receptors in healthy states and diseased states. These drug-like candidates are in turn exploited as radiolabeled probes for the imaging of chemokine receptors using noninvasive in vivo imaging, such as positron emission tomography (PET). Recent advances in the development of radiotracers for various chemokine receptors, particularly of CXCR4, CCR2, and CCR5, shed new light on chemokine-related cancer and cardiovascular research and the subsequent drug development. Here, we present the recent progress in PET radiotracer development for imaging of various chemokine receptors.
Collapse
Affiliation(s)
- Santosh R. Alluri
- University of Missouri Research Reactor, University of Missouri, Columbia, MO 65211, USA;
| | - Yusuke Higashi
- Department of Medicine, Tulane University, New Orleans, LA 70112, USA;
| | - Kun-Eek Kil
- University of Missouri Research Reactor, University of Missouri, Columbia, MO 65211, USA;
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO 65211, USA
- Correspondence: ; Tel.: +1-(573)-884-7885
| |
Collapse
|
50
|
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
|