1
|
Mirfakhraie N, Shoorei H, Abedpour N, Javanmard MZ. Co-treatment with bone marrow-derived mesenchymal stem cells and curcumin improved angiogenesis in myocardium in a rat model of MI. Mol Biol Rep 2024; 51:261. [PMID: 38302805 DOI: 10.1007/s11033-023-09180-z] [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: 09/19/2023] [Accepted: 12/18/2023] [Indexed: 02/03/2024]
Abstract
BACKGROUND The cardioprotective properties of mesenchymal stem cells and the therapeutic potential of curcumin (CUR) have been explored. Combining these approaches may enhance stem cell effectiveness and expedite healing. This study aimed to investigate the synergistic effects of co-treating bone marrow mesenchymal stem cells (BMSCs) with curcumin on vascular endothelial growth factor (VEGF) levels, in a rat model of myocardial ischemia (MI). METHODS AND RESULTS Sixty-five male rats were divided into four groups: G1 (healthy control), G2 (MI induced by isoproterenol hydrochloride), G3 (treated with BMSCs), and G4 (co-treated with curcumin and BMSCs). Blood and tissue samples were collected at specific time points (day 1, 7, 15 and 21) after MI induction. Serum levels of lactate dehydrogenase (LDH), creatine kinase (CK), cardiac troponin I (cTnI), aspartate aminotransferase (AST), CK-MB and VEGF were measured. VEGF mRNA and protein expression were evaluated using RT-qPCR and Western blot techniques. Histopathological assessments were performed using H&E staining and CD31 immunofluorescence staining. VEGF expression significantly increased on days 7 and 15 in the CUR-BMSCs group, peaking on day 7. Western blot analysis confirmed elevated VEGF protein expression on days 7 and 15 post-MI. ELISA results demonstrated increased serum VEGF levels on days 7 and 15, reaching the highest level on day 7 in CUR-BMSCs-treated animals. Treated groups showed lower levels of LDH, AST, CK, CK-MB and cTnI compared to the untreated MI group. H&E staining revealed improved myocardial structure, increased formation of new capillaries, in both treatment groups compared to the MI group. CONCLUSION Combining curcumin with BMSCs promotes angiogenesis in the infarcted myocardium after 15 days of MI induction. These findings suggest the potential of this combined therapy approach for enhancing cardiac healing and recovery.
Collapse
Affiliation(s)
- Niki Mirfakhraie
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Neda Abedpour
- Department of Anatomical Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Masoumeh Zirak Javanmard
- Department of Anatomical Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
2
|
Li B, Li Y, Chen S, Wang Y, Zheng Y. VEGF mimetic peptide-conjugated nanoparticles for magnetic resonance imaging and therapy of myocardial infarction. J Control Release 2023; 360:44-56. [PMID: 37330014 DOI: 10.1016/j.jconrel.2023.06.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 06/19/2023]
Abstract
To reduce the mortality of myocardial infarction (MI), accurate detection of the infarct and appropriate prevention against ischemia/reperfusion (I/R) induced cardiac dysfunction are highly desired. Considering that vascular endothelial growth factor (VEGF) receptors are overexpressed in the infarcted heart and VEGF mimetic peptide QK binds specifically to VEGF receptors and activates vascularization, the PEG-QK-modified, gadolinium-doped carbon dots (GCD-PEG-QK) were formulated. This research aims to investigate the magnetic resonance imaging (MRI) capability of GCD-PEG-QK on myocardial infarct and their therapeutic effect on I/R-induced myocardial injury. These multifunctional nanoparticles exhibited good colloidal stability, excellent fluorescent and magnetic property, and satisfactory biocompatibility. Intravenous injection of GCD-PEG-QK nanoparticles post myocardial I/R displayed accurate MRI of the infarct, enhanced efficacy of QK peptide on pro-angiogenesis, and amelioration of cardiac fibrosis, remodeling and dysfunction, probably via the improvement on QK's in vivo stability and MI-targeting. Collectively, the data suggested that this theranostic nanomedicine can realize precise MRI and effective therapy for acute MI in a non-invasive manner.
Collapse
Affiliation(s)
- Bing Li
- Department of Pharmacology, Capital Medical University, Beijing 100069, China
| | - Yingxu Li
- Department of Pharmacology, Capital Medical University, Beijing 100069, China
| | - Shuangling Chen
- Department of Chemical Biology, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
| | - Yuji Wang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
| | - Yuanyuan Zheng
- Department of Pharmacology, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
3
|
Neelamegam R, Chaly T, Dileep Kumar J. Radiosynthesis and in vivo imaging of [11C]BTFP, a potent inhibitor of VEGFR2. RESULTS IN CHEMISTRY 2022. [DOI: 10.1016/j.rechem.2022.100381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
4
|
Yaros K, Eksi B, Chandra A, Agusala K, Lehmann LH, Zaha Vlad G. Cardio-oncology imaging tools at the translational interface. J Mol Cell Cardiol 2022; 168:24-32. [DOI: 10.1016/j.yjmcc.2022.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 02/03/2022] [Accepted: 03/27/2022] [Indexed: 10/18/2022]
|
5
|
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
|
6
|
Tiwari A, Elgrably B, Saar G, Vandoorne K. Multi-Scale Imaging of Vascular Pathologies in Cardiovascular Disease. Front Med (Lausanne) 2022; 8:754369. [PMID: 35071257 PMCID: PMC8766766 DOI: 10.3389/fmed.2021.754369] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 12/13/2021] [Indexed: 12/28/2022] Open
Abstract
Cardiovascular disease entails systemic changes in the vasculature. The endothelial cells lining the blood vessels are crucial in the pathogenesis of cardiovascular disease. Healthy endothelial cells direct the blood flow to tissues as vasodilators and act as the systemic interface between the blood and tissues, supplying nutrients for vital organs, and regulating the smooth traffic of leukocytes into tissues. In cardiovascular diseases, when inflammation is sensed, endothelial cells adjust to the local or systemic inflammatory state. As the inflamed vasculature adjusts, changes in the endothelial cells lead to endothelial dysfunction, altered blood flow and permeability, expression of adhesion molecules, vessel wall inflammation, thrombosis, angiogenic processes, and extracellular matrix production at the endothelial cell level. Preclinical multi-scale imaging of these endothelial changes using optical, acoustic, nuclear, MRI, and multimodal techniques has progressed, due to technical advances and enhanced biological understanding on the interaction between immune and endothelial cells. While this review highlights biological processes that are related to changes in the cardiac vasculature during cardiovascular diseases, it also summarizes state-of-the-art vascular imaging techniques. The advantages and disadvantages of the different imaging techniques are highlighted, as well as their principles, methodologies, and preclinical and clinical applications with potential future directions. These multi-scale approaches of vascular imaging carry great potential to further expand our understanding of basic vascular biology, to enable early diagnosis of vascular changes and to provide sensitive diagnostic imaging techniques in the management of cardiovascular disease.
Collapse
Affiliation(s)
- Ashish Tiwari
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Betsalel Elgrably
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Galit Saar
- Biomedical Core Facility, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Katrien Vandoorne
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
7
|
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
|
8
|
The Role of VEGF Receptors as Molecular Target in Nuclear Medicine for Cancer Diagnosis and Combination Therapy. Cancers (Basel) 2021; 13:cancers13051072. [PMID: 33802353 PMCID: PMC7959315 DOI: 10.3390/cancers13051072] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/13/2021] [Accepted: 02/24/2021] [Indexed: 12/17/2022] Open
Abstract
Simple Summary The rapid development of diagnostic and therapeutic methods of the cancer treatment causes that these diseases are becoming better known and the fight against them is more and more effective. Substantial contribution in this development has nuclear medicine that enables very early cancer diagnosis and early start of the so-called targeted therapy. This therapeutic concept compared to the currently used chemotherapy, causes much fewer undesirable side effects, due to targeting a specific lesion in the body. This review article discusses the possible applications of radionuclide-labelled tracers (peptides, antibodies or synthetic organic molecules) that can visualise cancer cells through pathological blood vessel system in close tumour microenvironment. Hence, at a very early step of oncological disease, targeted therapy can involve in tumour formation and growth. Abstract One approach to anticancer treatment is targeted anti-angiogenic therapy (AAT) based on prevention of blood vessel formation around the developing cancer cells. It is known that vascular endothelial growth factor (VEGF) and vascular endothelial growth factor receptors (VEGFRs) play a pivotal role in angiogenesis process; hence, application of angiogenesis inhibitors can be an effective approach in anticancer combination therapeutic strategies. Currently, several types of molecules have been utilised in targeted VEGF/VEGFR anticancer therapy, including human VEGF ligands themselves and their derivatives, anti-VEGF or anti-VEGFR monoclonal antibodies, VEGF binding peptides and small molecular inhibitors of VEGFR tyrosine kinases. These molecules labelled with diagnostic or therapeutic radionuclides can become, respectively, diagnostic or therapeutic receptor radiopharmaceuticals. In targeted anti-angiogenic therapy, diagnostic radioagents play a unique role, allowing the determination of the emerging tumour, to monitor the course of treatment, to predict the treatment outcomes and, first of all, to refer patients for AAT. This review provides an overview of design, synthesis and study of radiolabelled VEGF/VEGFR targeting and imaging agents to date. Additionally, we will briefly discuss their physicochemical properties and possible application in combination targeted radionuclide tumour therapy.
Collapse
|
9
|
Hu K, Shang J, Xie L, Hanyu M, Zhang Y, Yang Z, Xu H, Wang L, Zhang MR. PET Imaging of VEGFR with a Novel 64Cu-Labeled Peptide. ACS OMEGA 2020; 5:8508-8514. [PMID: 32337411 PMCID: PMC7178340 DOI: 10.1021/acsomega.9b03953] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 03/12/2020] [Indexed: 05/11/2023]
Abstract
Vascular endothelial growth factor receptors (VEGFRs) are well recognized as significant biomarkers of tumor angiogenesis. Herein, we have developed a first-of-its-kind peptide-based VEGFR positron emission tomography (PET) tracer. The novel [64Cu]VEGF125-136 peptide possessed satisfactory radio-characteristics and showed good specificity for the visualization of VEGFR in various mouse models, in which the tumor-specific radioactivity uptake was highly correlated to the VEGFR expression level. Moreover, the tracer showed high tumor uptake (ca. 5.89 %ID/g at 20 min postinjection in B16F10 mice) and excellent pharmacokinetics, achieving the maximum imaging quality within 1 h after injection. These features convey [64Cu]VEGF125-136 as a promising, clinically translatable PET tracer for the imaging of tumor angiogenesis.
Collapse
Affiliation(s)
- Kuan Hu
- Department
of Advanced Nuclear Medicine Sciences, National
Institute of Radiological Sciences, National Institutes for Quantum
and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Jingjie Shang
- Center
of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine
and PET/CT-MRI Center, The First Affiliated
Hospital of Jinan University, Guangzhou 510630, China
| | - Lin Xie
- Department
of Advanced Nuclear Medicine Sciences, National
Institute of Radiological Sciences, National Institutes for Quantum
and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Masayuki Hanyu
- Department
of Advanced Nuclear Medicine Sciences, National
Institute of Radiological Sciences, National Institutes for Quantum
and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Yiding Zhang
- Department
of Advanced Nuclear Medicine Sciences, National
Institute of Radiological Sciences, National Institutes for Quantum
and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Zhimin Yang
- Department
of Advanced Nuclear Medicine Sciences, National
Institute of Radiological Sciences, National Institutes for Quantum
and Radiological Science and Technology, Chiba 263-8555, Japan
- Center
of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine
and PET/CT-MRI Center, The First Affiliated
Hospital of Jinan University, Guangzhou 510630, China
| | - Hao Xu
- Center
of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine
and PET/CT-MRI Center, The First Affiliated
Hospital of Jinan University, Guangzhou 510630, China
| | - Lu Wang
- Center
of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine
and PET/CT-MRI Center, The First Affiliated
Hospital of Jinan University, Guangzhou 510630, China
| | - Ming-Rong Zhang
- Department
of Advanced Nuclear Medicine Sciences, National
Institute of Radiological Sciences, National Institutes for Quantum
and Radiological Science and Technology, Chiba 263-8555, Japan
| |
Collapse
|
10
|
Hedhli J, Kim M, Knox HJ, Cole JA, Huynh T, Schuelke M, Dobrucki IT, Kalinowski L, Chan J, Sinusas AJ, Insana MF, Dobrucki LW. Imaging the Landmarks of Vascular Recovery. Am J Cancer Res 2020; 10:1733-1745. [PMID: 32042333 PMCID: PMC6993245 DOI: 10.7150/thno.36022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 08/31/2019] [Indexed: 12/25/2022] Open
Abstract
Background: Peripheral arterial disease (PAD) is a major worldwide health concern. Since the late 1990s therapeutic angiogenesis has been investigated as an alternative to traditional PAD treatments. Although positive preclinical results abound in the literature, the outcomes of human clinical trials have been discouraging. Among the challenges the field has faced has been a lack of standardization of the timings and measures used to validate new treatment approaches. Methods: In order to study the spatiotemporal dynamics of both perfusion and neovascularization in mice subjected to surgically-induced hindlimb ischemia (n= 30), we employed three label-free imaging modalities (a novel high-sensitivity ultrasonic Power Doppler methodology, laser speckle contrast, and photoacoustic imaging), as well as a tandem of radio-labeled molecular probes, 99mTc-NC100692 and 99mTc-BRU-5921 respectively, designed to detect two key modulators of angiogenic activity, αVβ3 and HIF-1α , via scintigraphic imaging. Results: The multimodal imaging strategy reveals a set of “landmarks”—key physiological and molecular events in the healing process—that can serve as a standardized framework for describing the impact of emerging PAD treatments. These landmarks span the entire process of neovascularization, beginning with the rapid decreases in perfusion and oxygenation associated with ligation surgery, extending through pro-angiogenic changes in gene expression driven by the master regulator HIF-1α , and ultimately leading to complete functional revascularization of the affected tissues. Conclusions: This study represents an important step in the development of multimodal non-invasive imaging strategies for vascular research; the combined results offer more insight than can be gleaned through any of the individual imaging methods alone. Researchers adopting similar imaging strategies and will be better able to describe changes in the onset, duration, and strength of each of the landmarks of vascular recovery, yielding greater biological insight, and enabling more comprehensive cross-study comparisons. Perhaps most important, this study paves the road for more efficient translation of PAD research; emerging experimental treatments can be more effectively assessed and refined at the preclinical stage, ultimately leading to better next-generation therapies.
Collapse
|
11
|
In vivo imaging of TGFβ signalling components using positron emission tomography. Drug Discov Today 2019; 24:2258-2272. [DOI: 10.1016/j.drudis.2019.08.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 08/01/2019] [Accepted: 08/28/2019] [Indexed: 12/21/2022]
|
12
|
Boutagy NE, Feher A, Alkhalil I, Umoh N, Sinusas AJ. Molecular Imaging of the Heart. Compr Physiol 2019; 9:477-533. [PMID: 30873600 DOI: 10.1002/cphy.c180007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Multimodality cardiovascular imaging is routinely used to assess cardiac function, structure, and physiological parameters to facilitate the diagnosis, characterization, and phenotyping of numerous cardiovascular diseases (CVD), as well as allows for risk stratification and guidance in medical therapy decision-making. Although useful, these imaging strategies are unable to assess the underlying cellular and molecular processes that modulate pathophysiological changes. Over the last decade, there have been great advancements in imaging instrumentation and technology that have been paralleled by breakthroughs in probe development and image analysis. These advancements have been merged with discoveries in cellular/molecular cardiovascular biology to burgeon the field of cardiovascular molecular imaging. Cardiovascular molecular imaging aims to noninvasively detect and characterize underlying disease processes to facilitate early diagnosis, improve prognostication, and guide targeted therapy across the continuum of CVD. The most-widely used approaches for preclinical and clinical molecular imaging include radiotracers that allow for high-sensitivity in vivo detection and quantification of molecular processes with single photon emission computed tomography and positron emission tomography. This review will describe multimodality molecular imaging instrumentation along with established and novel molecular imaging targets and probes. We will highlight how molecular imaging has provided valuable insights in determining the underlying fundamental biology of a wide variety of CVDs, including: myocardial infarction, cardiac arrhythmias, and nonischemic and ischemic heart failure with reduced and preserved ejection fraction. In addition, the potential of molecular imaging to assist in the characterization and risk stratification of systemic diseases, such as amyloidosis and sarcoidosis will be discussed. © 2019 American Physiological Society. Compr Physiol 9:477-533, 2019.
Collapse
Affiliation(s)
- Nabil E Boutagy
- Department of Medicine, Yale Translational Research Imaging Center, Yale University School of Medicine, Section of Cardiovascular Medicine, New Haven, Connecticut, USA
| | - Attila Feher
- Department of Medicine, Yale Translational Research Imaging Center, Yale University School of Medicine, Section of Cardiovascular Medicine, New Haven, Connecticut, USA
| | - Imran Alkhalil
- Department of Medicine, Yale Translational Research Imaging Center, Yale University School of Medicine, Section of Cardiovascular Medicine, New Haven, Connecticut, USA
| | - Nsini Umoh
- Department of Medicine, Yale Translational Research Imaging Center, Yale University School of Medicine, Section of Cardiovascular Medicine, New Haven, Connecticut, USA
| | - Albert J Sinusas
- Department of Medicine, Yale Translational Research Imaging Center, Yale University School of Medicine, Section of Cardiovascular Medicine, New Haven, Connecticut, USA.,Yale University School of Medicine, Department of Radiology and Biomedical Imaging, New Haven, Connecticut, USA
| |
Collapse
|
13
|
Molecular imaging of cardiac remodelling after myocardial infarction. Basic Res Cardiol 2018; 113:10. [PMID: 29344827 PMCID: PMC5772148 DOI: 10.1007/s00395-018-0668-z] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 11/17/2017] [Accepted: 01/08/2018] [Indexed: 02/06/2023]
Abstract
Myocardial infarction and subsequent heart failure is a major health burden associated with significant mortality and morbidity in western societies. The ability of cardiac tissue to recover after myocardial infarction is affected by numerous complex cellular and molecular pathways. Unbalance or failure of these pathways can lead to adverse remodelling of the heart and poor prognosis. Current clinical cardiac imaging modalities assess anatomy, perfusion, function, and viability of the myocardium, yet do not offer any insight into the specific molecular pathways involved in the repair process. Novel imaging techniques allow visualisation of these molecular processes and may have significant diagnostic and prognostic values, which could aid clinical management. Single photon-emission tomography, positron-emission tomography, and magnetic resonance imaging are used to visualise various aspects of these molecular processes. Imaging probes are usually attached to radioisotopes or paramagnetic nanoparticles to specifically target biological processes such as: apoptosis, necrosis, inflammation, angiogenesis, and scar formation. Although the results from preclinical studies are promising, translating this work to a clinical environment in a valuable and cost-effective way is extremely challenging. Extensive evaluation evidence of diagnostic and prognostic values in multi-centre clinical trials is still required.
Collapse
|
14
|
Chistiakov DA, Melnichenko AA, Myasoedova VA, Grechko AV, Orekhov AN. Role of lipids and intraplaque hypoxia in the formation of neovascularization in atherosclerosis. Ann Med 2017; 49:661-677. [PMID: 28797175 DOI: 10.1080/07853890.2017.1366041] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
According to the current paradigm, chronic vascular inflammation plays a central role in the pathogenesis of atherosclerosis. The plaque progression is typically completed with rupture and subsequent acute cardiovascular complications. Previously, the role of adventitial vasa vasorum in atherogenesis was underestimated. However, investigators then revealed that vasa vasorum neovascularization can be observed when no clinical manifestation of atherosclerosis is present. Vasa vasorum is involved in various proatherogenic processes such as intimal accumulation of inflammatory leukocytes, intimal thickening, necrotic core formation, intraplaque haemorrhage, lesion rupture and atherothrombosis. Due to the destabilizing action of the intraplaque microenvironment, lesional vasa vasorum neovessels experience serious defects and abnormalities during development that leads to their immaturity, fragility and leakage. Indeed, intraplaque neovessels are a main cause of intraplaque haemorrhage. Visualization techniques showed that presence of neovascularization/haemorrhage can serve as a good indicator of lesion instability and higher risk of rupture. Vasa vasorum density is a strong predictor of acute cardiovascular events such as sudden death, myocardial infarction and stroke. At present, arterial vasa vasorum neovascularization is under intensive investigation along with development of therapeutic tools focused on the control of formation of vasa vasorum neovessels in order to prevent plaque haemorrhage/rupture and thromboembolism. KEY MESSAGE Neovascularization plays an important role in atherosclerosis, being involved in unstable plaque formation. Presence of neovascularization and haemorrhage indicates plaque instability and risk of rupture. Various imaging techniques are available to study neovascularization.
Collapse
Affiliation(s)
- Dimitry A Chistiakov
- a Department of Neurochemistry, Division of Basic and Applied Neurobiology , Serbsky Federal Medical Research Center of Psychiatry and Narcology , Moscow , Russia
| | - Alexandra A Melnichenko
- b Laboratory of Angiopathology , Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences , Moscow , Russia
| | - Veronika A Myasoedova
- b Laboratory of Angiopathology , Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences , Moscow , Russia
| | - Andrey V Grechko
- c Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology , Moscow , Russia
| | - Alexander N Orekhov
- b Laboratory of Angiopathology , Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences , Moscow , Russia.,d Institute for Atherosclerosis Research, Skolkovo Innovative Center , Moscow , Russia
| |
Collapse
|
15
|
Li Z, Gupte AA, Zhang A, Hamilton DJ. Pet Imaging and its Application in Cardiovascular Diseases. Methodist Debakey Cardiovasc J 2017; 13:29-33. [PMID: 28413580 DOI: 10.14797/mdcj-13-1-29] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death worldwide and represent a great challenge for modern research and medicine. Despite advances in preventing and treating CVD over the decades, there remains an urgent need to develop sensitive and safe methods for early detection and personalized treatment. With refinements of molecular imaging technologies such as positron emission tomography (PET), noninvasive imaging of CVDs is experiencing impressive progress in both preclinical and clinical settings. In this review, we summarize advances in cardiovascular PET imaging, highlight the latest development of CVD imaging probes, and illustrate the potential for individualized therapy based on metabolic phenotype.
Collapse
Affiliation(s)
- Zheng Li
- Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas
| | - Anisha A Gupte
- Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas
| | - Anjun Zhang
- Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas
| | - Dale J Hamilton
- Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas
| |
Collapse
|
16
|
Paschali AN, Nekolla SG, Evangelou K, Cook GJ, Anagnostopoulos CD. One Coin, No Need to Flip: Shared PET Targets in Cancer and Coronary Artery Disease. AJR Am J Roentgenol 2017; 208:434-445. [PMID: 27897437 DOI: 10.2214/ajr.16.16599] [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] [Indexed: 11/18/2022]
Abstract
OBJECTIVE The purposes of this article are to review the common biologic features of cancer and coronary artery disease assessed with PET tracers, focusing on those already used in the clinic and those with translational potential, and to discuss the current value and expected contribution of PET in diagnosis, risk stratification, and treatment monitoring. CONCLUSION PET using a wide variety of radiotracers enhances understanding of pathophysiologic changes shared by cancer and coronary artery disease, helps establish an accurate diagnosis, and aids in prognostic assessment and management decisions. It is likely that with the evolution of therapeutic strategies for blocking the development and progression of both diseases and with the introduction of novel, specific ligands in clinical practice, PET will play an ever stronger role in diagnosis, risk stratification, and monitoring of therapy.
Collapse
Affiliation(s)
- Anna N Paschali
- 1 Department of Nuclear Medicine, Theagenion Cancer Hospital, Thessaloniki, Greece
| | - Stephen G Nekolla
- 2 Nuklearmedizinische Klinik und Poliklinik, Klinikum Rechts der Isar der Technischen Universitaet München, München, Germany
| | | | - Gary J Cook
- 4 PET Imaging Centre, St Thomas' Hospital, London, UK
| | - Constantinos D Anagnostopoulos
- 5 PET/CT Department and MicroPET/CT Unit, Center for Experimental Surgery, Clinical and Translational Research, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou St, 115 27 Athens, Greece
| |
Collapse
|
17
|
SPECT and PET imaging of angiogenesis and arteriogenesis in pre-clinical models of myocardial ischemia and peripheral vascular disease. Eur J Nucl Med Mol Imaging 2016; 43:2433-2447. [PMID: 27517840 PMCID: PMC5095166 DOI: 10.1007/s00259-016-3480-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 07/28/2016] [Indexed: 01/03/2023]
Abstract
Purpose The extent of neovascularization determines the clinical outcome of coronary artery disease and other occlusive cardiovascular disorders. Monitoring of neovascularization is therefore highly important. This review article will elaborately discuss preclinical studies aimed at validating new nuclear angiogenesis and arteriogenesis tracers. Additionally, we will briefly address possible obstacles that should be considered when designing an arteriogenesis radiotracer. Methods A structured medline search was the base of this review, which gives an overview on different radiopharmaceuticals that have been evaluated in preclinical models. Results Neovascularization is a collective term used to indicate different processes such as angiogenesis and arteriogenesis. However, while it is assumed that sensitive detection through nuclear imaging will facilitate translation of successful therapeutic interventions in preclinical models to the bedside, we still lack specific tracers for neovascularization imaging. Most nuclear imaging research to date has focused on angiogenesis, leaving nuclear arteriogenesis imaging largely overlooked. Conclusion Although angiogenesis is the process which is best understood, there is no scarcity in theoretical targets for arteriogenesis imaging.
Collapse
|
18
|
Molecular Imaging of Angiogenesis and Vascular Remodeling in Cardiovascular Pathology. J Clin Med 2016; 5:jcm5060057. [PMID: 27275836 PMCID: PMC4929412 DOI: 10.3390/jcm5060057] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 05/19/2016] [Accepted: 05/31/2016] [Indexed: 12/20/2022] Open
Abstract
Angiogenesis and vascular remodeling are involved in a wide array of cardiovascular diseases, from myocardial ischemia and peripheral arterial disease, to atherosclerosis and aortic aneurysm. Molecular imaging techniques to detect and quantify key molecular and cellular players in angiogenesis and vascular remodeling (e.g., vascular endothelial growth factor and its receptors, αvβ3 integrin, and matrix metalloproteinases) can advance vascular biology research and serve as clinical tools for early diagnosis, risk stratification, and selection of patients who would benefit most from therapeutic interventions. To target these key mediators, a number of molecular imaging techniques have been developed and evaluated in animal models of angiogenesis and vascular remodeling. This review of the state of the art molecular imaging of angiogenesis and vascular (and valvular) remodeling, will focus mostly on nuclear imaging techniques (positron emission tomography and single photon emission tomography) that offer high potential for clinical translation.
Collapse
|
19
|
Park S, Lee J, Jo MH, Na JH, Park SG, Jang HK, Kang SW, Kim JH, Kim BS, Park JH, Kwon IC, Ryu JH, Kim K. In vivo monitoring of angiogenesis in a mouse hindlimb ischemia model using fluorescent peptide-based probes. Amino Acids 2016; 48:1641-54. [DOI: 10.1007/s00726-016-2225-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 03/25/2016] [Indexed: 12/13/2022]
|
20
|
Luo H, England CG, Graves SA, Sun H, Liu G, Nickles RJ, Cai W. PET Imaging of VEGFR-2 Expression in Lung Cancer with 64Cu-Labeled Ramucirumab. J Nucl Med 2016; 57:285-90. [PMID: 26541778 PMCID: PMC4738068 DOI: 10.2967/jnumed.115.166462] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 10/21/2015] [Indexed: 12/16/2022] Open
Abstract
UNLABELLED Lung cancer accounts for 17% of cancer-related deaths worldwide, and most patients present with locally advanced or metastatic disease. Novel PET imaging agents for assessing vascular endothelial growth factor receptor-2 (VEGFR-2) expression can be used for detecting VEGFR-2-positive malignancies and subsequent monitoring of therapeutic response to VEGFR-2-targeted therapies. Here, we report the synthesis and characterization of an antibody-based imaging agent for PET imaging of VEGFR-2 expression in vivo. METHODS Ramucirumab (named RamAb), a fully humanized IgG1 monoclonal antibody, was conjugated to 2-S-(4-isothiocyanatobenzyl)-1,4,7-triazacyclononane-1,4,7-triacetic acid (p-SCN-Bn-NOTA) and labeled with (64)Cu. Flow cytometry analysis and microscopy studies were performed to compare the VEGFR-2 binding affinity of RamAb and NOTA-RamAb. PET imaging and biodistribution studies were performed in nude mice bearing HCC4006 and A549 xenograft tumors. Ex vivo histopathology was performed to elucidate the expression patterns of VEGFR-2 in different tissues and organs to validate in vivo results. RESULTS Flow cytometry examination revealed the specific binding capacity of fluorescein isothiocyanate-RamAb to VEGFR-2, and no difference in VEGFR-2 binding affinity was seen between RamAb and NOTA-RamAb. After being labeled with (64)Cu, PET imaging revealed specific and prominent uptake of (64)Cu-NOTA-RamAb in VEGFR-2-positive HCC4006 tumors (9.4 ± 0.5 percentage injected dose per gram at 48 h after injection; n = 4) and significantly lower uptake in VEGFR-2-negative A549 tumors (4.3 ± 0.2 percentage injected dose per gram at 48 h after injection; n = 3). Blocking experiments revealed significantly lower uptake in HCC4006 tumors, along with histology analysis, further confirming the VEGFR-2 specificity of (64)Cu-NOTA-RamAb. CONCLUSION This study provides initial evidence that (64)Cu-NOTA-RamAb can function as a PET imaging agent for visualizing VEGFR-2 expression in vivo, which may also find potential applications in monitoring the treatment response of VEGFR-2-targeted cancer therapy.
Collapse
Affiliation(s)
- Haiming Luo
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Christopher G England
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin; and
| | - Stephen A Graves
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin; and
| | - Haiyan Sun
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Glenn Liu
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| | - Robert J Nickles
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin; and
| | - Weibo Cai
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin; and University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| |
Collapse
|
21
|
Vonk Noordegraaf A, Haddad F, Bogaard HJ, Hassoun PM. Noninvasive imaging in the assessment of the cardiopulmonary vascular unit. Circulation 2015; 131:899-913. [PMID: 25753343 DOI: 10.1161/circulationaha.114.006972] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Anton Vonk Noordegraaf
- From Pulmonary Diseases (A.V.N., J.H.B.) and Physics and Medical Technology (A.V.N.), Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands; Division of Cardiovascular Medicine, Department of Medicine and Stanford Cardiovascular Institute, Stanford University, Palo Alto, CA (F.H.); and Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, MD (P.M.H.).
| | - Francois Haddad
- From Pulmonary Diseases (A.V.N., J.H.B.) and Physics and Medical Technology (A.V.N.), Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands; Division of Cardiovascular Medicine, Department of Medicine and Stanford Cardiovascular Institute, Stanford University, Palo Alto, CA (F.H.); and Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, MD (P.M.H.)
| | - Harm J Bogaard
- From Pulmonary Diseases (A.V.N., J.H.B.) and Physics and Medical Technology (A.V.N.), Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands; Division of Cardiovascular Medicine, Department of Medicine and Stanford Cardiovascular Institute, Stanford University, Palo Alto, CA (F.H.); and Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, MD (P.M.H.)
| | - Paul M Hassoun
- From Pulmonary Diseases (A.V.N., J.H.B.) and Physics and Medical Technology (A.V.N.), Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands; Division of Cardiovascular Medicine, Department of Medicine and Stanford Cardiovascular Institute, Stanford University, Palo Alto, CA (F.H.); and Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, MD (P.M.H.)
| |
Collapse
|
22
|
Xu J, Lu X, Shi GP. Vasa vasorum in atherosclerosis and clinical significance. Int J Mol Sci 2015; 16:11574-608. [PMID: 26006236 PMCID: PMC4463718 DOI: 10.3390/ijms160511574] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 05/11/2015] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease that leads to several acute cardiovascular complications with poor prognosis. For decades, the role of the adventitial vasa vasorum (VV) in the initiation and progression of atherosclerosis has received broad attention. The presence of VV neovascularization precedes the apparent symptoms of clinical atherosclerosis. VV also mediates inflammatory cell infiltration, intimal thickening, intraplaque hemorrhage, and subsequent atherothrombosis that results in stroke or myocardial infarction. Intraplaque neovessels originating from VV can be immature and hence susceptible to leakage, and are thus regarded as the leading cause of intraplaque hemorrhage. Evidence supports VV as a new surrogate target of atherosclerosis evaluation and treatment. This review provides an overview into the relationship between VV and atherosclerosis, including the anatomy and function of VV, the stimuli of VV neovascularization, and the available underlying mechanisms that lead to poor prognosis. We also summarize translational researches on VV imaging modalities and potential therapies that target VV neovascularization or its stimuli.
Collapse
Affiliation(s)
- Junyan Xu
- Second Clinical Medical College, Zhujiang Hospital and Southern Medical University, Guangzhou 510280, China.
| | - Xiaotong Lu
- Second Clinical Medical College, Zhujiang Hospital and Southern Medical University, Guangzhou 510280, China.
| | - Guo-Ping Shi
- Second Clinical Medical College, Zhujiang Hospital and Southern Medical University, Guangzhou 510280, China.
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
23
|
Lin JB, Phillips EH, Riggins TE, Sangha GS, Chakraborty S, Lee JY, Lycke RJ, Hernandez CL, Soepriatna AH, Thorne BRH, Yrineo AA, Goergen CJ. Imaging of small animal peripheral artery disease models: recent advancements and translational potential. Int J Mol Sci 2015; 16:11131-77. [PMID: 25993289 PMCID: PMC4463694 DOI: 10.3390/ijms160511131] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 03/10/2015] [Indexed: 12/11/2022] Open
Abstract
Peripheral artery disease (PAD) is a broad disorder encompassing multiple forms of arterial disease outside of the heart. As such, PAD development is a multifactorial process with a variety of manifestations. For example, aneurysms are pathological expansions of an artery that can lead to rupture, while ischemic atherosclerosis reduces blood flow, increasing the risk of claudication, poor wound healing, limb amputation, and stroke. Current PAD treatment is often ineffective or associated with serious risks, largely because these disorders are commonly undiagnosed or misdiagnosed. Active areas of research are focused on detecting and characterizing deleterious arterial changes at early stages using non-invasive imaging strategies, such as ultrasound, as well as emerging technologies like photoacoustic imaging. Earlier disease detection and characterization could improve interventional strategies, leading to better prognosis in PAD patients. While rodents are being used to investigate PAD pathophysiology, imaging of these animal models has been underutilized. This review focuses on structural and molecular information and disease progression revealed by recent imaging efforts of aortic, cerebral, and peripheral vascular disease models in mice, rats, and rabbits. Effective translation to humans involves better understanding of underlying PAD pathophysiology to develop novel therapeutics and apply non-invasive imaging techniques in the clinic.
Collapse
Affiliation(s)
- Jenny B Lin
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, Room 3025, West Lafayette, IN 47907, USA.
| | - Evan H Phillips
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, Room 3025, West Lafayette, IN 47907, USA.
| | - Ti'Air E Riggins
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, Room 3025, West Lafayette, IN 47907, USA.
| | - Gurneet S Sangha
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, Room 3025, West Lafayette, IN 47907, USA.
| | - Sreyashi Chakraborty
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47907, USA.
| | - Janice Y Lee
- Psychological Sciences, Purdue University, West Lafayette, IN 47907, USA.
| | - Roy J Lycke
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, Room 3025, West Lafayette, IN 47907, USA.
| | - Clarissa L Hernandez
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, Room 3025, West Lafayette, IN 47907, USA.
| | - Arvin H Soepriatna
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, Room 3025, West Lafayette, IN 47907, USA.
| | - Bradford R H Thorne
- School of Sciences, Neuroscience, Purdue University, West Lafayette, IN 47907, USA.
| | - Alexa A Yrineo
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, Room 3025, West Lafayette, IN 47907, USA.
| | - Craig J Goergen
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, Room 3025, West Lafayette, IN 47907, USA.
| |
Collapse
|
24
|
Stacy MR, Paeng JC, Sinusas AJ. The role of molecular imaging in the evaluation of myocardial and peripheral angiogenesis. Ann Nucl Med 2015; 29:217-23. [PMID: 25750124 PMCID: PMC4661208 DOI: 10.1007/s12149-015-0961-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 02/26/2015] [Indexed: 11/28/2022]
Abstract
Angiogenesis, or the formation of new microvasculature, is a physiological process that may occur in the setting of chronic tissue ischemia and can play an important role in improving tissue perfusion and blood flow following myocardial infarction or in the presence of peripheral vascular disease (PVD). Molecular imaging of angiogenesis within the cardiovascular system is a developing field of study. Targeted imaging of angiogenesis has the potential for non-invasive assessment of the underlying molecular signaling events associated with the angiogenic process and, when applied in conjunction with physiological perfusion imaging, may be utilized to predict and evaluate clinical outcomes in the setting of ischemic heart disease or PVD. This review discusses the developing radiotracer-based imaging techniques and technology currently in use that possess potential for clinical translation, with specific focus on PET and SPECT imaging of myocardial and peripheral angiogenesis.
Collapse
Affiliation(s)
- Mitchel R Stacy
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale University School of Medicine, Dana-3, P.O. Box 208017, New Haven, CT, 06520, USA
| | | | | |
Collapse
|
25
|
Cardiac MRI and PET Scanning in Right Ventricular Failure. THE RIGHT VENTRICLE IN HEALTH AND DISEASE 2015. [DOI: 10.1007/978-1-4939-1065-6_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
26
|
Advanced tracers in PET imaging of cardiovascular disease. BIOMED RESEARCH INTERNATIONAL 2014; 2014:504532. [PMID: 25389529 PMCID: PMC4214169 DOI: 10.1155/2014/504532] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 07/07/2014] [Accepted: 08/08/2014] [Indexed: 02/04/2023]
Abstract
Cardiovascular disease is the leading cause of death worldwide. Molecular imaging with targeted tracers by positron emission tomography (PET) allows for the noninvasive detection and characterization of biological changes at the molecular level, leading to earlier disease detection, objective monitoring of therapies, and better prognostication of cardiovascular diseases progression. Here we review, the current role of PET in cardiovascular disease, with emphasize on tracers developed for PET imaging of cardiovascular diseases.
Collapse
|
27
|
|
28
|
van de Veerdonk MC, Marcus JT, Bogaard HJ, Vonk Noordegraaf A. State of the art: advanced imaging of the right ventricle and pulmonary circulation in humans (2013 Grover Conference series). Pulm Circ 2014; 4:158-68. [PMID: 25006434 DOI: 10.1086/675978] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 12/03/2013] [Indexed: 12/27/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease characterized by remodeling and vasoconstriction of the pulmonary vasculature, ultimately leading to right ventricular (RV) failure and death. Recent developments in echocardiography, cardiovascular magnetic resonance imaging, computed tomography, and positron emission tomography allow advanced, noninvasive, in vivo assessment of the RV and have contributed to the identification of risk factors, prognostic factors, and monitoring of therapeutic responses in patients with PAH. Although far from reaching its future potential, these techniques have not only provided global RV assessment but also allowed evaluation of changes in cellular and molecular tissue processes, such as metabolism, oxygen balance and ischemia, angiogenesis, and apoptosis. Integrated application of these techniques could provide full insights into the different pathophysiological aspects of a failing RV in the setting of PAH. Recent advances in hybrid imaging have implemented simultaneous measurements of myocardial and vascular interactions and will be one of the most important potential future developments.
Collapse
Affiliation(s)
- Mariëlle C van de Veerdonk
- Pulmonary Diseases, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands
| | - J Tim Marcus
- Physics and Medical Technology, ICaR-VU, VU University Medical Center, Amsterdam, The Netherlands
| | - Harm-Jan Bogaard
- Pulmonary Diseases, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands
| | - Anton Vonk Noordegraaf
- Pulmonary Diseases, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
29
|
Li F, Jiang S, Zu Y, Lee DY, Li Z. A Tyrosine Kinase Inhibitor–Based High-Affinity PET Radiopharmaceutical Targets Vascular Endothelial Growth Factor Receptor. J Nucl Med 2014; 55:1525-31. [DOI: 10.2967/jnumed.114.138925] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
30
|
Abstract
Nuclear imaging techniques that include positron emission tomography (PET) and single-photon computed tomography have found great success in the clinic because of their inherent high sensitivity. Radionuclide imaging is the most popular form of imaging to be used for molecular imaging in oncology. While many types of molecules have been used for radionuclide-based molecular imaging, there has been a great interest in developing newer nanomaterials for use in clinic, especially for cancer diagnosis and treatment. Nanomaterials have unique physical properties which allow them to be used as imaging probes to locate and identify cancerous lesions. Over the past decade, a great number of nanoparticles have been developed for radionuclide imaging of cancer. This chapter reviews the different kinds of nanomaterials, both organic and inorganic, which are currently being researched for as potential agents for nuclear imaging of variety of cancers. Several radiolabeled multifunctional nanocarriers have been extremely successful for the detection of cancer in preclinical models. So far, significant progress has been achieved in nanoparticle structure design, in vitro/in vivo trafficking, and in vivo fate mapping by using PET. There is a great need for the development of newer nanoparticles, which improve active targeting and quantify new biomarkers for early disease detection and possible prevention of cancer.
Collapse
|
31
|
Orbay H, Zhang Y, Valdovinos HF, Song G, Hernandez R, Theuer CP, Hacker TA, Nickles RJ, Cai W. Positron emission tomography imaging of CD105 expression in a rat myocardial infarction model with (64)Cu-NOTA-TRC105. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2013; 4:1-9. [PMID: 24380040 PMCID: PMC3867724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Accepted: 11/17/2013] [Indexed: 06/03/2023]
Abstract
Biological changes following myocardial infarction (MI) lead to increased secretion of angiogenic factors that subsequently stimulate the formation of new blood vessels as a compensatory mechanism to reverse ischemia. The goal of this study was to assess the role of CD105 expression during MI-induced angiogenesis by positron emission tomography (PET) imaging using (64)Cu-labeled TRC105, an anti-CD105 monoclonal antibody. MI was induced by ligation of the left anterior descending (LAD) artery in female rats. Echocardiography and (18)F-fluoro-2-deoxy-D-glucose ((18)F-FDG) PET scans were performed on post-operative day 3 to confirm the presence of MI in the infarct group and intact heart in the sham group, respectively. Ischemia-induced angiogenesis was non-invasively monitored with (64)Cu-NOTA-TRC105 (an extensively validated PET tracer in our previous studies) PET on post-operative days 3, 10, and 17. Tracer uptake in the infarct zone was highest on day 3 following MI, which was significantly higher than that in the sham group (1.41 ± 0.45 %ID/g vs 0.57 ± 0.07 %ID/g; n=3, p<0.05). Subsequently, tracer uptake in the infarct zone decreased over time to the background level on day 17, whereas tracer uptake in the heart of sham rats remained low at all time points examined. Histopathology documented increased CD105 expression following MI, which corroborated in vivo findings. This study indicated that PET imaging of CD105 can be a useful tool for MI-related research, which can potentially improve MI patient management in the future upon clinical translation of the optimized PET tracers.
Collapse
Affiliation(s)
- Hakan Orbay
- Department of Radiology, University of Wisconsin - MadisonWI, USA
| | - Yin Zhang
- Department of Medical Physics, University of Wisconsin - MadisonWI, USA
| | | | - Guoqing Song
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin - MadisonWI, USA
| | - Reinier Hernandez
- Department of Medical Physics, University of Wisconsin - MadisonWI, USA
| | | | - Timothy A Hacker
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin - MadisonWI, USA
| | - Robert J Nickles
- Department of Medical Physics, University of Wisconsin - MadisonWI, USA
| | - Weibo Cai
- Department of Radiology, University of Wisconsin - MadisonWI, USA
- Department of Medical Physics, University of Wisconsin - MadisonWI, USA
- University of Wisconsin Carbone Cancer CenterMadison, WI, USA
| |
Collapse
|
32
|
Harms HJ, van de Veerdonk MC, Lammertsma AA, Vonk Noordegraaf A, Bogaard HJ. Pulmonary vascular remodeling and right heart failure in pulmonary hypertension: future role of positron emission tomography in decoding the enigma. TRANSLATIONAL RESPIRATORY MEDICINE 2013; 1:16. [PMID: 27234397 PMCID: PMC4715170 DOI: 10.1186/2213-0802-1-16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 11/18/2013] [Indexed: 11/23/2022]
Abstract
Whereas the insights into the cellular and molecular mechanisms of pulmonary arterial hypertension (PAH) and associated right heart failure have increased in recent years, there is a lack of clinical tools to assess the pathobiological mechanisms in patients. Positron emission tomography (PET) provides an array of new possibilities to image and quantify relevant disease processes, including proliferation, angiogenesis, matrix remodeling, shifts in metabolism and neurohormonal signaling. Here we describe the first studies which were conducted to image pulmonary vascular remodeling and right heart failure in vivo and discuss additional targets for imaging which hold great promise for future use in PAH patients.
Collapse
Affiliation(s)
- Hendrik J Harms
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Mariëlle C van de Veerdonk
- Department of Pulmonary Medicine, VU University Medical Center, PO Box 7057, Amsterdam, MB, 1007, The Netherlands
| | - Adriaan A Lammertsma
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Anton Vonk Noordegraaf
- Department of Pulmonary Medicine, VU University Medical Center, PO Box 7057, Amsterdam, MB, 1007, The Netherlands
| | - Harm Jan Bogaard
- Department of Pulmonary Medicine, VU University Medical Center, PO Box 7057, Amsterdam, MB, 1007, The Netherlands.
| |
Collapse
|
33
|
Jivraj N, Phinikaridou A, Shah AM, Botnar RM. Molecular imaging of myocardial infarction. Basic Res Cardiol 2013; 109:397. [PMID: 24322905 DOI: 10.1007/s00395-013-0397-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 11/14/2013] [Accepted: 11/27/2013] [Indexed: 11/29/2022]
Abstract
Myocardial infarction (MI), and subsequent heart failure, remains a major healthcare problem in the western and developing world and leads to substantial morbidity and mortality. After MI, the ability of the myocardium to recover is closely associated with a complex immune response that often leads to adverse remodeling of the ventricle, and poor prognosis. Currently used clinical imaging modalities allow the assessment of anatomy, perfusion, function, and viability but do not provide insights into specific biological processes. In contrast, novel non-invasive imaging methods, using targeted imaging agents, allow imaging of the molecular processes underlying the post-MI immune cell response, and subsequent remodeling. Therefore, this may have significant diagnostic, prognostic, and therapeutic value, and may help to improve our understanding of post-infarct remodeling, in vivo. Imaging modalities such as magnetic resonance imaging, single-photon emission computed tomography, and positron emission tomography have been used in concert with radiolabelled and (super) paramagnetic probes to image each phase of the immune response. These probes, which target apoptosis, necrosis, neutrophils, monocytes, enzymes, angiogenesis, extracellular matrix, and scar formation have been assessed and validated pre-clinically. Translating this work to the bedside in a cost-effective, clinically beneficial manner remains a significant challenge. This article reviews these new imaging techniques as well as the corresponding pathophysiology.
Collapse
Affiliation(s)
- Naheed Jivraj
- Division of Imaging Sciences and Biomedical Engineering, King's College London, St. Thomas' Hospital, 4th Floor, Lambeth Wing, London, SE1 7EH, UK,
| | | | | | | |
Collapse
|
34
|
Böning G, Todica A, Vai A, Lehner S, Xiong G, Mille E, Ilhan H, la Fougère C, Bartenstein P, Hacker M. Erroneous cardiac ECG-gated PET list-mode trigger events can be retrospectively identified and replaced by an offline reprocessing approach: first results in rodents. Phys Med Biol 2013; 58:7937-59. [PMID: 24165267 DOI: 10.1088/0031-9155/58/22/7937] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The assessment of left ventricular function, wall motion and myocardial viability using electrocardiogram (ECG)-gated [(18)F]-FDG positron emission tomography (PET) is widely accepted in human and in preclinical small animal studies. The nonterminal and noninvasive approach permits repeated in vivo evaluations of the same animal, facilitating the assessment of temporal changes in disease or therapy response. Although well established, gated small animal PET studies can contain erroneous gating information, which may yield to blurred images and false estimation of functional parameters. In this work, we present quantitative and visual quality control (QC) methods to evaluate the accuracy of trigger events in PET list-mode and physiological data. Left ventricular functional analysis is performed to quantify the effect of gating errors on the end-systolic and end-diastolic volumes, and on the ejection fraction (EF). We aim to recover the cardiac functional parameters by the application of the commonly established heart rate filter approach using fixed ranges based on a standardized population. In addition, we propose a fully reprocessing approach which retrospectively replaces the gating information of the PET list-mode file with appropriate list-mode decoding and encoding software. The signal of a simultaneously acquired ECG is processed using standard MATLAB vector functions, which can be individually adapted to reliably detect the R-peaks. Finally, the new trigger events are inserted into the PET list-mode file. A population of 30 mice with various health statuses was analyzed and standard cardiac parameters such as mean heart rate (119 ms ± 11.8 ms) and mean heart rate variability (1.7 ms ± 3.4 ms) derived. These standard parameter ranges were taken into account in the QC methods to select a group of nine optimal gated and a group of eight sub-optimal gated [(18)F]-FDG PET scans of mice from our archive. From the list-mode files of the optimal gated group, we randomly deleted various fractions (5% to 60%) of contained trigger events to generate a corrupted group. The filter approach was capable to correct the corrupted group and yield functional parameters with no significant difference to the optimal gated group. We successfully demonstrated the potential of the fully reprocessing approach by applying it to the sub-optimal group, where the functional parameters were significantly improved after reprocessing (mean EF from 41% ± 16% to 60% ± 13%). When applied to the optimal gated group the fully reprocessing approach did not alter the functional parameters significantly (mean EF from 64% ± 8% to 64 ± 7%). This work presents methods to determine and quantify erroneous gating in small animal gated [(18)F]-FDG PET scans. We demonstrate the importance of a quality check for cardiac triggering contained in PET list-mode data and the benefit of optionally reprocessing the fully recorded physiological information to retrospectively modify or fully replace the cardiac triggering in PET list-mode data. We aim to provide a preliminary guideline of how to proceed in the presence of errors and demonstrate that offline reprocessing by filtering erroneous trigger events and retrospective gating by ECG processing is feasible. Future work will focus on the extension by additional QC methods, which may exploit the amplitude of trigger events and ECG signal by means of pattern recognition. Furthermore, we aim to transfer the proposed QC methods and the fully reprocessing approach to human myocardial PET/CT.
Collapse
Affiliation(s)
- Guido Böning
- Department of Nuclear Medicine, Ludwig-Maximilians University of Munich, Munich, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Kang CM, Koo HJ, Choe YS, Choi JY, Lee KH, Kim BT. ⁶⁸Ga-NODAGA-VEGF₁₂₁ for in vivo imaging of VEGF receptor expression. Nucl Med Biol 2013; 41:51-7. [PMID: 24183611 DOI: 10.1016/j.nucmedbio.2013.09.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 09/12/2013] [Accepted: 09/13/2013] [Indexed: 11/30/2022]
Abstract
PURPOSE Vascular endothelial growth factor (VEGF) is a crucial regulator of angiogenesis. In this study, we labeled VEGF₁₂₁ with (68)Ga using a hydrophilic chelating agent, NODAGA and evaluated the resulting (68)Ga-NODAGA-VEGF₁₂₁ for in vivo imaging of VEGF receptor (VEGFR) expression. METHODS NODAGA-VEGF₁₂₁ was prepared and its binding affinity for VEGFR2 was measured using (125)I-VEGF₁₂₁. (68)Ga-NODAGA-VEGF₁₂₁ was prepared by labeling NODAGA-VEGF₁₂₁ with (68)GaCl3 followed by purification using a PD-10 column. Human aortic endothelial cell (HAEC) binding studies of (68)Ga-NODAGA-VEGF₁₂₁ were performed at 37°C for 4 h. MicroPET imaging followed by biodistribution studies were performed in U87MG tumor-bearing mice injected with (68)Ga-NODAGA-VEGF₁₂₁. Immunofluorescence staining of the tumor tissues was performed to verify VEGFR2 expression. RESULTS Binding affinity of NODAGA-VEGF₁₂₁ for VEGFR2 was found to be comparable to that of VEGF₁₂₁. (68)Ga-NODAGA-VEGF₁₂₁ was prepared in 47.8% yield with specific activity of 3.4 GBq/mg. (68)Ga-NODAGA-VEGF₁₂₁ was avidly taken up by HAECs with a time-dependent increase from 9.88 %ID at 1 h to 20.86 %ID at 4h. MicroPET imaging of mice demonstrated high liver and spleen uptake with clear visualization of tumor at 1h after injection. ROI analysis of tumors revealed 2.53 ± 0.11 %ID/g at 4 h after injection. In the blocking study, tumor uptake was inhibited by 29% at 4 h. Subsequent biodistribution studies demonstrated tumor uptake of 2.38 ± 0.15 %ID/g. Immunofluorescence staining of the tumor tissues displayed high level of VEGFR2 expression. CONCLUSIONS These results demonstrate that (68)Ga-NODAGA-VEGF₁₂₁ led to VEGFR-specific distribution in U87MG tumor-bearing mice. This study also suggests that altered physicochemical properties of VEGF₁₂₁ after radiolabeling may affect biodistribution of the radiolabeled VEGF₁₂₁.
Collapse
Affiliation(s)
- Choong Mo Kang
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Ilwon-dong, Kangnam-ku, Seoul 135-710, Korea; Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 135-710, Korea
| | | | | | | | | | | |
Collapse
|
36
|
A vascular endothelial growth factor 121 (VEGF121)-based dual PET/optical probe for in vivo imaging of VEGF receptor expression. Biomaterials 2013; 34:6839-45. [DOI: 10.1016/j.biomaterials.2013.05.051] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 05/23/2013] [Indexed: 11/18/2022]
|
37
|
Jenkins WSA, Chin C, Rudd JHF, Newby DE, Dweck MR. What can we learn about valvular heart disease from PET/CT? Future Cardiol 2013; 9:657-67. [DOI: 10.2217/fca.13.47] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Valvular heart disease is a major cause of morbidity and mortality, and with an aging population, its prevalence is increasing. Here, we review the evolving use of positron emission tomography/computed tomography in valvular heart disease, with particular focus on calcific aortic stenosis and infective endocarditis. In principle, the activity of any pathological process can be studied, as long as an appropriate radiotracer can be developed. We will review some of the early data using established tracers in the above and other conditions, providing discussion as to the future research and clinical roles of these techniques. Furthermore, we will discuss the potential impact of novel tracers that are currently under development or testing in preclinical models. It is hoped that such advanced imaging might improve the diagnosis, treatment and outlook for patients with valvular heart disease.
Collapse
Affiliation(s)
- William SA Jenkins
- Centre for Cardiovascular Science, University of Edinburgh, Little France Crescent, Edinburgh, UK
| | - Calvin Chin
- Centre for Cardiovascular Science, University of Edinburgh, Little France Crescent, Edinburgh, UK
- National Heart Center Singapore, SingHealth, Singapore
| | - James HF Rudd
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - David E Newby
- Centre for Cardiovascular Science, University of Edinburgh, Little France Crescent, Edinburgh, UK
| | - Marc R Dweck
- Centre for Cardiovascular Science, University of Edinburgh, Little France Crescent, Edinburgh, UK
| |
Collapse
|
38
|
|
39
|
Fernández-Friera L, García-Álvarez A, Ibáñez B. Imagining the future of diagnostic imaging. ACTA ACUST UNITED AC 2012; 66:134-43. [PMID: 24775390 DOI: 10.1016/j.rec.2012.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 10/01/2012] [Indexed: 01/21/2023]
Abstract
Cardiovascular imaging has become essential to achieving a better understanding of cardiovascular diseases. Due to the advent of new technology and the refinement of existing technologies, imaging's role has extended into the biological, functional, and hemodynamic diagnosis of multiple pathophysiologic processes. Current and future trends in cardiovascular imaging will focus on improving early diagnosis of vascular disease, so as to be able to promote cardiovascular health, and on its development as a useful tool in clinical decision-making. Imaging is also increasingly used to quantify the effect of novel therapies. The rapid development of molecular imaging and fusion imaging techniques improves our understanding of cardiovascular processes from the molecular and cellular points of view and makes it possible to design and test new preventive interventions. The proliferation and integration of imaging techniques in different clinical areas and their role in "translational imaging" plays an important part in the implementation of personalized therapeutic and preventive management strategies for patients with cardiovascular disease.
Collapse
Affiliation(s)
- Leticia Fernández-Friera
- Centro Nacional de Investigaciones Cardiovasculares (CNIC) - Imaging in Experimental Cardiology Laboratory (IExC Lab), Madrid, Spain; Servicio de Cardiología, Hospital Universitario Montepríncipe, Madrid, Spain
| | - Ana García-Álvarez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC) - Imaging in Experimental Cardiology Laboratory (IExC Lab), Madrid, Spain; Servicio de Cardiología, Hospital Clínic, Barcelona, Spain
| | - Borja Ibáñez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC) - Imaging in Experimental Cardiology Laboratory (IExC Lab), Madrid, Spain; Instituto Cardiovascular, Hospital Clínico San Carlos, Madrid, Spain.
| |
Collapse
|
40
|
Kang CM, Kim SM, Koo HJ, Yim MS, Lee KH, Ryu EK, Choe YS. In vivo characterization of 68Ga-NOTA-VEGF 121 for the imaging of VEGF receptor expression in U87MG tumor xenograft models. Eur J Nucl Med Mol Imaging 2012; 40:198-206. [PMID: 23096079 DOI: 10.1007/s00259-012-2266-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Accepted: 09/26/2012] [Indexed: 11/27/2022]
Abstract
PURPOSE Vascular endothelial growth factor receptors (VEGFRs) are associated with tumor growth and induction of tumor angiogenesis and are known to be overexpressed in various human tumors. In the present study, we prepared and evaluated (68)Ga-1,4,7-triazacyclononane-1,4,7-triacetic acid-benzyl (NOTA)-VEGF(121) as a positron emission tomography (PET) radioligand for the in vivo imaging of VEGFR expression. METHODS (68)Ga-NOTA-VEGF(121) was prepared by conjugation of VEGF(121) and p-SCN-NOTA, followed by radiolabeling with (68)GaCl(3) and then purification using a PD-10 column. Human aortic endothelial cell (HAEC) binding of (68)Ga-NOTA-VEGF(121) was measured as a function of time. MicroPET and biodistribution studies of U87MG tumor xenografted mice were performed at 1, 2, and 4 h after injection of (68)Ga-NOTA-VEGF(121). The tumor tissues were then sectioned and subjected to immunostaining. RESULTS The decay-corrected radiochemical yield of (68)Ga-NOTA-VEGF(121) was 40 ± 4.5 % and specific activity was 243.1 ± 104.6 GBq/μmol (8.6 ± 3.7 GBq/mg). (68)Ga-NOTA-VEGF(121) was avidly taken up by HAECs in a time-dependent manner, and the uptake was blocked either by 32 % with VEGF(121) or by 49 % with VEGFR2 antibody at 4 h post-incubation. In microPET images of U87MG tumor xenografted mice, radioactivity was accumulated in tumors (2.73±0.32 %ID/g at 2 h), and the uptake was blocked by 40 % in the presence of VEGF(121). In biodistribution studies, tumor uptake (1.84±0.14 %ID/g at 2 h) was blocked with VEGF(121) at a similar level (52 %) to that of microPET images. Immunostaining analysis of U87MG tumor tissues obtained after the microPET imaging showed high levels of VEGFR2 expression. CONCLUSION These results demonstrate that (68)Ga-NOTA-VEGF(121) has potential for the in vivo imaging of VEGFR expression. In addition, our results also suggest that the in vivo characteristics of radiolabeled VEGF depend on the properties of the radioisotope and the chelator used.
Collapse
Affiliation(s)
- Choong Mo Kang
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Ilwon-dong, Kangnam-ku, Seoul 135-710, South Korea
| | | | | | | | | | | | | |
Collapse
|
41
|
Phinikaridou A, Andia ME, Shah AM, Botnar RM. Advances in molecular imaging of atherosclerosis and myocardial infarction: shedding new light on in vivo cardiovascular biology. Am J Physiol Heart Circ Physiol 2012; 303:H1397-410. [PMID: 23064836 DOI: 10.1152/ajpheart.00583.2012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Molecular imaging of the cardiovascular system heavily relies on the development of new imaging probes and technologies to facilitate visualization of biological processes underlying or preceding disease. Molecular imaging is a highly active research discipline that has seen tremendous growth over the past decade. It has broadened our understanding of oncologic, neurologic, and cardiovascular diseases by providing new insights into the in vivo biology of disease progression and therapeutic interventions. As it allows for the longitudinal evaluation of biological processes, it is ideally suited for monitoring treatment response. In this review, we will concentrate on the major accomplishments and advances in the field of molecular imaging of atherosclerosis and myocardial infarction with a special focus on magnetic resonance imaging.
Collapse
Affiliation(s)
- Alkystis Phinikaridou
- Division of Imaging Science and Biomedical Engineering, King's College London, United Kingdom.
| | | | | | | |
Collapse
|
42
|
Individualizing therapy of monoclonal antibodies and fusion proteins: emerging potential in the age of personalized medicine. Ther Deliv 2012; 2:369-81. [PMID: 22834007 DOI: 10.4155/tde.10.101] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Personalized medicine is an emerging and promising alternative to standard therapy regimens with the potential to significantly influence therapeutic interventions for many diseases. An extensive literature review of studies that focused on pharmacogenomics of monoclonal antibodies (mAbs) and immunoglobulin-containing fusion proteins (igFPs) was conducted. A comprehensive survey of the US FDA-approved labels revealed that pharmacogenomics information has also been incorporated into the label of some mAbs to guide therapy. In addition, treatment-emergent adverse events for mAbs and igFPs were analyzed that showed an association with the drugs' individual mechanism of action as well as molecular nature. The identification of the signaling pathways linked to the specific target of each mAb or igFP may help accelerate clinical successes in predicting and managing treatment-associated severe adverse events in individual patients. Incorporating pharmacogenomics into drug development of mAbs and igFPs will improve treatment efficacy, and may allow prediction of adverse events. Thus, a promising future of personalized medicine for these therapeutics is predicted.
Collapse
|
43
|
Prabhakaran J, Arango V, Majo VJ, Simpson NR, Kassir SA, Underwood MD, Polavarapu H, Bruce JN, Canoll P, Mann JJ, Kumar JSD. Synthesis and in vitro evaluation of [18F](R)-FEPAQ: a potential PET ligand for VEGFR2. Bioorg Med Chem Lett 2012; 22:5104-7. [PMID: 22749281 PMCID: PMC4818572 DOI: 10.1016/j.bmcl.2012.05.099] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 05/25/2012] [Accepted: 05/29/2012] [Indexed: 12/19/2022]
Abstract
Synthesis and in vitro evaluation of [(18)F](R)-N-(4-bromo-2-fluorophenyl)-7-((1-(2-fluoroethyl)piperidin-3-yl)methoxy)-6-methoxyquinazolin-4-amine ((R)-[(18)F]FEPAQ or [(18)F]1), a potential imaging agent for the VEGFR2, using phosphor image autoradiography are described. Synthesis of 2, the desfluoroethyl precursor for (R)-FEPAQ was achieved from t-butyl 3-(hydroxymethyl)piperidine-1-carboxylate (3) in five steps and in 50% yield. [(18)F]1 was synthesized by reaction of sodium salt of compound 2 with [(18)F]fluoroethyl tosylate in DMSO. The yield of [(18)F]1 was 20% (EOS based on [(18)F]F(-)) with >99% radiochemical purity and specific activity of 1-2 Ci/μmol (n=10). The total synthesis time was 75 min. The radiotracer selectively labeled VEGFR2 in slide-mounted sections of human brain and higher binding was found in surgically removed human glioblastoma sections as demonstrated by in vitro phosphor imager studies. These findings suggest [(18)F]1 may be a promising radiotracer for imaging VEGFR2 in brain using PET.
Collapse
Affiliation(s)
- Jaya Prabhakaran
- Division of Molecular Imaging and Neuropathology, Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, New York, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Kataoka Y, Uno K, Puri R, Nicholls SJ. Current imaging modalities for atherosclerosis. Expert Rev Cardiovasc Ther 2012; 10:457-71. [PMID: 22458579 DOI: 10.1586/erc.12.28] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Atherosclerotic disease is responsible for nearly half of all deaths in the western world. During the past three decades, considerable efforts have been made towards detection and assessment of atherosclerosis plaques in various vascular beds using different imaging techniques. Recently, both noninvasive and invasive modalities have frequently been used to refine cardiovascular risk assessment in high-risk individuals, to evaluate the natural history of atheroma burden and to reveal the impact of anti-atherosclerotic medical therapies on disease progression. In this review, we provide an overview of the currently available imaging modalities. This article will underscore arterial wall imaging to assess the impact of medical therapies on atherosclerosis and to develop the effective therapeutic strategies, resulting in the prevention of cardiovascular complications.
Collapse
Affiliation(s)
- Yu Kataoka
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | | | | | | |
Collapse
|
45
|
Abstract
Although FDG PET and PET/CT have a well established role in the management of most cancer patients, they also have some limitations. For the last 15-20 years a growing number of non-FDG PET tracers have been used in research. Many of these new PET tracers are being investigated for the non-invasive assessment of different biologic functions in cancer cells. This unique information should contribute to making personalized cancer therapy a reality. This paper reviews the non-FDG PET tracers that are most likely to find clinical application, some of them in the near future.
Collapse
|
46
|
Zhang J, Razavian M, Tavakoli S, Nie L, Tellides G, Backer JM, Backer MV, Bender JR, Sadeghi MM. Molecular imaging of vascular endothelial growth factor receptors in graft arteriosclerosis. Arterioscler Thromb Vasc Biol 2012; 32:1849-55. [PMID: 22723442 DOI: 10.1161/atvbaha.112.252510] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Vascular endothelial growth factor (VEGF) signaling plays a key role in the pathogenesis of vascular remodeling, including graft arteriosclerosis. Graft arteriosclerosis is the major cause of late organ failure in cardiac transplantation. We used molecular near-infrared fluorescent imaging with an engineered Cy5.5-labeled single-chain VEGF tracer (scVEGF/Cy) to detect VEGF receptors and vascular remodeling in human coronary artery grafts by molecular imaging. METHODS AND RESULTS VEGF receptor specificity of probe uptake was shown by flow cytometry in endothelial cells. In severe combined immunodeficiency mice, transplantation of human coronary artery segments into the aorta followed by adoptive transfer of allogeneic human peripheral blood mononuclear cells led to significant neointima formation in the grafts over a period of 4 weeks. Near-infrared fluorescent imaging of transplant recipients at 4 weeks demonstrated focal uptake of scVEGF/Cy in remodeling artery grafts. Uptake specificity was demonstrated using an inactive homolog of scVEGF/Cy. scVEGF/Cy uptake predominantly localized in the neointima of remodeling coronary arteries and correlated with VEGF receptor-1 but not VEGF receptor-2 expression. There was a significant correlation between scVEGF/Cy uptake and transplanted artery neointima area. CONCLUSIONS Molecular imaging of VEGF receptors may provide a noninvasive tool for detection of graft arteriosclerosis in solid organ transplantation.
Collapse
Affiliation(s)
- Jiasheng Zhang
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Molecular Imaging of Left Ventricular Remodeling. CURRENT CARDIOVASCULAR IMAGING REPORTS 2012. [DOI: 10.1007/s12410-012-9137-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
48
|
Abstract
Despite significant advancements in medical and device-based therapies, cardiovascular disease remains the number one cause of death in the United States. Early detection of atherosclerosis, prevention of myocardial infarction and sudden cardiac death, and modulation of adverse ventricular remodeling still remain elusive goals. Molecular imaging focuses on identifying critical cellular and molecular targets and therefore plays an integral role in understanding these biologic processes in vivo. Because many imaging targets are upregulated before irreversible tissue damage occurs, early detection could ultimately lead to development of novel, preventive therapeutic strategies. This review addresses recent work on radionuclide imaging of cardiovascular inflammation, infection, and infarct healing. We further discuss opportunities provided by multimodality approaches such as PET/MRI and PET/optical imaging.
Collapse
Affiliation(s)
- Maulik D Majmudar
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | | |
Collapse
|
49
|
Stacy MR, Maxfield MW, Sinusas AJ. Targeted molecular imaging of angiogenesis in PET and SPECT: a review. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2012; 85:75-86. [PMID: 22461745 PMCID: PMC3313541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Over the past few decades, there have been significant advancements in the imaging techniques of positron emission tomography (PET) and single photon emission tomography (SPECT). These changes have allowed for the targeted imaging of cellular processes and the development of hybrid imaging systems (e.g., SPECT/CT and PET/CT), which provide both functional and structural images of biological systems. One area that has garnered particular attention is angiogenesis as it relates to ischemic heart disease and limb ischemia. Though the aforementioned techniques have benefits and consequences, they enable scientists and clinicians to identify regions that are vulnerable to or have been exposed to ischemic injury via non-invasive means. This literature review highlights the advancements in molecular imaging techniques and specific probes as they pertain to the process of angiogenesis in cardiovascular disease.
Collapse
Affiliation(s)
- Mitchel R. Stacy
- Section of Cardiovascular Medicine, Department of
Internal Medicine, Yale School of Medicine, New Haven, Connecticut,To whom all correspondence should be
addressed: Mitchel R. Stacy, Nuclear Cardiology, 3 FMP, PO Box 208017, New
Haven, CT 06520-8017, Tel: 203-737-5917; Fax: 203-737-1030;
| | - Mark W. Maxfield
- Department of Surgery, Yale School of Medicine, New
Haven, Connecticut
| | - Albert J. Sinusas
- Section of Cardiovascular Medicine, Department of
Internal Medicine, Yale School of Medicine, New Haven, Connecticut,Department of Diagnostic Radiology, Yale School of
Medicine, New Haven, Connecticut
| |
Collapse
|
50
|
Zhang Y, Hong H, Engle JW, Yang Y, Barnhart TE, Cai W. Positron Emission Tomography and Near-Infrared Fluorescence Imaging of Vascular Endothelial Growth Factor with Dual-Labeled Bevacizumab. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2012; 2:1-13. [PMID: 22229128 PMCID: PMC3249831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 12/12/2011] [Indexed: 05/31/2023]
Abstract
The pivotal role of vascular endothelial growth factor (VEGF) in cancer is underscored by the approval of bevacizumab (Bev, a humanized anti-VEGF monoclonal antibody) for first line treatment of cancer patients. The aim of this study was to develop a dual-labeled Bev for both positron emission tomography (PET) and near-infrared fluorescence (NIRF) imaging of VEGF. Bev was conjugated to a NIRF dye (i.e. 800CW) and 2-S-(4-isothiocyanatobenzyl)-1,4,7-triazacyclononane-1,4,7-triacetic acid (p-SCN-Bn-NOTA) before (64)Cu-labeling. Flow cytometry analysis of U87MG human glioblastoma cells revealed no difference in VEGF binding affinity/specificity between Bev and NOTA-Bev-800CW. (64)Cu-labeling of NOTA-Bev-800CW was achieved with high yield. Serial PET imaging of U87MG tumor-bearing female nude mice revealed that tumor uptake of (64)Cu-NOTA-Bev-800CW was 4.6 ± 0.7, 16.3 ± 1.6, 18.1 ± 1.4 and 20.7 ± 3.7 %ID/g at 4, 24, 48 and 72 h post-injection respectively (n = 4), corroborated by in vivo/ex vivo NIRF imaging and biodistribution studies. Tumor uptake as measured by ex vivo NIRF imaging had a good linear correlation with the %ID/g values obtained from PET (R(2) = 0.93). Blocking experiments and histology both confirmed the VEGF specificity of (64)Cu-NOTA-Bev-800CW. The persistent, prominent, and VEGF-specific uptake of (64)Cu-NOTA-Bev-800CW in the tumor, observed by both PET and NIRF imaging, warrants further investigation and future clinical translation of such Bev-based imaging agents.
Collapse
Affiliation(s)
- Yin Zhang
- Department of Medical Physics, University of Wisconsin - MadisonMadison, WI, USA
| | - Hao Hong
- Department of Radiology, University of Wisconsin - MadisonMadison, WI, USA
| | - Jonathan W Engle
- Department of Medical Physics, University of Wisconsin - MadisonMadison, WI, USA
| | - Yunan Yang
- Department of Radiology, University of Wisconsin - MadisonMadison, WI, USA
| | - Todd E Barnhart
- Department of Medical Physics, University of Wisconsin - MadisonMadison, WI, USA
| | - Weibo Cai
- Department of Medical Physics, University of Wisconsin - MadisonMadison, WI, USA
- Department of Radiology, University of Wisconsin - MadisonMadison, WI, USA
- University of Wisconsin Carbone Cancer CenterMadison, WI, USA
| |
Collapse
|