1
|
Esposito TVF, Rodríguez-Rodríguez C, Blackadar C, Kłodzińska S, Mørck Nielsen H, Saatchi K, Häfeli UO. Biodistribution of the cationic host defense peptide LL-37 using SPECT/CT. Eur J Pharm Biopharm 2024; 202:114398. [PMID: 38972467 DOI: 10.1016/j.ejpb.2024.114398] [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: 04/02/2024] [Revised: 07/02/2024] [Accepted: 07/04/2024] [Indexed: 07/09/2024]
Abstract
Human cathelicidin LL-37, a cationic host defense peptide (CHDP), has several important physiological roles, including antimicrobial activity, immune modulation, and wound healing, and is a being investigated as a therapeutic candidate for several indications. While the effects of endogenously produced LL-37 are well studied, the biodistribution of exogenously administered LL-37 are less known. Here we assess the biodistribution of a gallium-67 labeled variant of LL-37 using nuclear imaging techniques over a 48 h period in healthy mice. When administered as an intravenous bolus just over 20 µg, the LL-37-based radiotracer was rapidly cleared from the blood, largely by the liver, while an appreciable fraction of the dose temporarily distributed to the lungs. When administered subcutaneously at the same dose level, the radiotracer was absorbed systemically following a two-phase kinetic model and was predominately cleared renally. Uptake into sites rich in immune cells, such as the lymph nodes and the spleen, was observed for both routes of administration. Scans of free gallium-67 were also performed as controls. Important preclinical insights into the biodistribution of exogenously administered LL-37 were gained from this study, which can aid in the understanding of this and related cationic host-defense peptides.
Collapse
Affiliation(s)
- Tullio V F Esposito
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada.
| | - Cristina Rodríguez-Rodríguez
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada; Department of Physics and Astronomy, Faculty of Science, University of British Columbia, Vancouver, Canada
| | - Colin Blackadar
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada
| | - Sylvia Kłodzińska
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hanne Mørck Nielsen
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Katayoun Saatchi
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada.
| | - Urs O Häfeli
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada; Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
2
|
Maier A, Teunissen AJP, Nauta SA, Lutgens E, Fayad ZA, van Leent MMT. Uncovering atherosclerotic cardiovascular disease by PET imaging. Nat Rev Cardiol 2024; 21:632-651. [PMID: 38575752 PMCID: PMC11324396 DOI: 10.1038/s41569-024-01009-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/04/2024] [Indexed: 04/06/2024]
Abstract
Assessing atherosclerosis severity is essential for precise patient stratification. Specifically, there is a need to identify patients with residual inflammation because these patients remain at high risk of cardiovascular events despite optimal management of cardiovascular risk factors. Molecular imaging techniques, such as PET, can have an essential role in this context. PET imaging can indicate tissue-based disease status, detect early molecular changes and provide whole-body information. Advances in molecular biology and bioinformatics continue to help to decipher the complex pathogenesis of atherosclerosis and inform the development of imaging tracers. Concomitant advances in tracer synthesis methods and PET imaging technology provide future possibilities for atherosclerosis imaging. In this Review, we summarize the latest developments in PET imaging techniques and technologies for assessment of atherosclerotic cardiovascular disease and discuss the relationship between imaging readouts and transcriptomics-based plaque phenotyping.
Collapse
Affiliation(s)
- Alexander Maier
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Abraham J P Teunissen
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sheqouia A Nauta
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Esther Lutgens
- Cardiovascular Medicine and Immunology, Experimental Cardiovascular Immunology Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Zahi A Fayad
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mandy M T van Leent
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
3
|
Shen L, Bi Y, Yu J, Zhong Y, Chen W, Zhao Z, Ding J, Shu G, Chen M, Lu C, Ji J. The biological applications of near-infrared optical nanomaterials in atherosclerosis. J Nanobiotechnology 2024; 22:478. [PMID: 39135099 PMCID: PMC11320980 DOI: 10.1186/s12951-024-02703-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 07/05/2024] [Indexed: 08/15/2024] Open
Abstract
PURPOSE OF REVIEW Atherosclerosis, a highly pathogenic and lethal disease, is difficult to locate accurately via conventional imaging because of its scattered and deep lesions. However, second near-infrared (NIR-II) nanomaterials show great application potential in the tracing of atherosclerotic plaques due to their excellent penetration and angiographic capabilities. RECENT FINDINGS With the development of nanotechnology, among many nanomaterials available for the visual diagnosis and treatment of cardiovascular diseases, optical nanomaterials provide strong support for various biomedical applications because of their advantages, such as noninvasive, nondestructive and molecular component imaging. Among optical nanomaterials of different wavelengths, NIR-II-range (900 ~ 1700 nm) nanomaterials have been gradually applied in the visual diagnosis and treatment of atherosclerosis and other vascular diseases because of their deep biological tissue penetration and limited background interference. This review explored in detail the prospects and challenges of the biological imaging and clinical application of NIR-II nanomaterials in treating atherosclerosis.
Collapse
Affiliation(s)
- Lin Shen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Yanran Bi
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Junchao Yu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Yi Zhong
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Weiqian Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Zhongwei Zhao
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Jiayi Ding
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Gaofeng Shu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Minjiang Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Chenying Lu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China
| | - Jiansong Ji
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China.
- Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui, 323000, China.
| |
Collapse
|
4
|
Liu Y, Jiang Z, Yang X, Wang Y, Yang B, Fu Q. Engineering Nanoplatforms for Theranostics of Atherosclerotic Plaques. Adv Healthc Mater 2024; 13:e2303612. [PMID: 38564883 DOI: 10.1002/adhm.202303612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 03/28/2024] [Indexed: 04/04/2024]
Abstract
Atherosclerotic plaque formation is considered the primary pathological mechanism underlying atherosclerotic cardiovascular diseases, leading to severe cardiovascular events such as stroke, acute coronary syndromes, and even sudden cardiac death. Early detection and timely intervention of plaques are challenging due to the lack of typical symptoms in the initial stages. Therefore, precise early detection and intervention play a crucial role in risk stratification of atherosclerotic plaques and achieving favorable post-interventional outcomes. The continuously advancing nanoplatforms have demonstrated numerous advantages including high signal-to-noise ratio, enhanced bioavailability, and specific targeting capabilities for imaging agents and therapeutic drugs, enabling effective visualization and management of atherosclerotic plaques. Motivated by these superior properties, various noninvasive imaging modalities for early recognition of plaques in the preliminary stage of atherosclerosis are comprehensively summarized. Additionally, several therapeutic strategies are proposed to enhance the efficacy of treating atherosclerotic plaques. Finally, existing challenges and promising prospects for accelerating clinical translation of nanoplatform-based molecular imaging and therapy for atherosclerotic plaques are discussed. In conclusion, this review provides an insightful perspective on the diagnosis and therapy of atherosclerotic plaques.
Collapse
Affiliation(s)
- Yuying Liu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Zeyu Jiang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Xiao Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Bin Yang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Qinrui Fu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| |
Collapse
|
5
|
Toner YC, Prévot G, van Leent MMT, Munitz J, Oosterwijk R, Verschuur AVD, van Elsas Y, Peric V, Maas RJF, Ranzenigo A, Morla-Folch J, Wang W, Umali M, de Dreu A, Fernandes JC, Sullivan NAT, Maier A, Mason C, Reiner T, Fayad ZA, Mulder WJM, Teunissen AJP, Pérez-Medina C. Macrophage PET imaging in mouse models of cardiovascular disease and cancer with an apolipoprotein-inspired radiotracer. NPJ IMAGING 2024; 2:12. [PMID: 38765879 PMCID: PMC11096117 DOI: 10.1038/s44303-024-00009-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 02/06/2024] [Indexed: 05/22/2024]
Abstract
Macrophages are key inflammatory mediators in many pathological conditions, including cardiovascular disease (CVD) and cancer, the leading causes of morbidity and mortality worldwide. This makes macrophage burden a valuable diagnostic marker and several strategies to monitor these cells have been reported. However, such strategies are often high-priced, non-specific, invasive, and/or not quantitative. Here, we developed a positron emission tomography (PET) radiotracer based on apolipoprotein A1 (ApoA1), the main protein component of high-density lipoprotein (HDL), which has an inherent affinity for macrophages. We radiolabeled an ApoA1-mimetic peptide (mA1) with zirconium-89 (89Zr) to generate a lipoprotein-avid PET probe (89Zr-mA1). We first characterized 89Zr-mA1's affinity for lipoproteins in vitro by size exclusion chromatography. To study 89Zr-mA1's in vivo behavior and interaction with endogenous lipoproteins, we performed extensive studies in wildtype C57BL/6 and Apoe-/- hypercholesterolemic mice. Subsequently, we used in vivo PET imaging to study macrophages in melanoma and myocardial infarction using mouse models. The tracer's cell specificity was assessed by histology and mass cytometry (CyTOF). Our data show that 89Zr-mA1 associates with lipoproteins in vitro. This is in line with our in vivo experiments, in which we observed longer 89Zr-mA1 circulation times in hypercholesterolemic mice compared to C57BL/6 controls. 89Zr-mA1 displayed a tissue distribution profile similar to ApoA1 and HDL, with high kidney and liver uptake as well as substantial signal in the bone marrow and spleen. The tracer also accumulated in tumors of melanoma-bearing mice and in the ischemic myocardium of infarcted animals. In these sites, CyTOF analyses revealed that natZr-mA1 was predominantly taken up by macrophages. Our results demonstrate that 89Zr-mA1 associates with lipoproteins and hence accumulates in macrophages in vivo. 89Zr-mA1's high uptake in these cells makes it a promising radiotracer for non-invasively and quantitatively studying conditions characterized by marked changes in macrophage burden.
Collapse
Affiliation(s)
- Yohana C. Toner
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Geoffrey Prévot
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Mandy M. T. van Leent
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Jazz Munitz
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Roderick Oosterwijk
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Anna Vera D. Verschuur
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Yuri van Elsas
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Vedran Peric
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Rianne J. F. Maas
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Anna Ranzenigo
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Judit Morla-Folch
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - William Wang
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Martin Umali
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Anne de Dreu
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Jessica Chimene Fernandes
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Nathaniel A. T. Sullivan
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Alexander Maier
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Cardiology and Angiology, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christian Mason
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY USA
- Department of Radiology, Weill Cornell Medical College, New York, NY USA
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Zahi A. Fayad
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Willem J. M. Mulder
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Abraham J. P. Teunissen
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Carlos Pérez-Medina
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| |
Collapse
|
6
|
Guo J, Wang H, Li Y, Zhu S, Hu H, Gu Z. Nanotechnology in coronary heart disease. Acta Biomater 2023; 171:37-67. [PMID: 37714246 DOI: 10.1016/j.actbio.2023.09.011] [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: 05/22/2023] [Revised: 08/17/2023] [Accepted: 09/08/2023] [Indexed: 09/17/2023]
Abstract
Coronary heart disease (CHD) is one of the major causes of death and disability worldwide, especially in low- and middle-income countries and among older populations. Conventional diagnostic and therapeutic approaches have limitations such as low sensitivity, high cost and side effects. Nanotechnology offers promising alternative strategies for the diagnosis and treatment of CHD by exploiting the unique properties of nanomaterials. In this review, we use bibliometric analysis to identify research hotspots in the application of nanotechnology in CHD and provide a comprehensive overview of the current state of the art. Nanomaterials with enhanced imaging and biosensing capabilities can improve the early detection of CHD through advanced contrast agents and high-resolution imaging techniques. Moreover, nanomaterials can facilitate targeted drug delivery, tissue engineering and modulation of inflammation and oxidative stress, thus addressing multiple aspects of CHD pathophysiology. We discuss the application of nanotechnology in CHD diagnosis (imaging and sensors) and treatment (regulation of macrophages, cardiac repair, anti-oxidative stress), and provide insights into future research directions and clinical translation. This review serves as a valuable resource for researchers and clinicians seeking to harness the potential of nanotechnology in the management of CHD. STATEMENT OF SIGNIFICANCE: Coronary heart disease (CHD) is the one of leading cause of death and disability worldwide. Nanotechnology offers new strategies for diagnosing and treating CHD by exploiting the unique properties of nanomaterials. This review uses bibliometric analysis to uncover research trends in the use of nanotechnology for CHD. We discuss the potential of nanomaterials for early CHD detection through advanced imaging and biosensing, targeted drug delivery, tissue engineering, and modulation of inflammation and oxidative stress. We also offer insights into future research directions and potential clinical applications. This work aims to guide researchers and clinicians in leveraging nanotechnology to improve CHD patient outcomes and quality of life.
Collapse
Affiliation(s)
- Junsong Guo
- Academician Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China; Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Hao Wang
- Academician Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China; Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Ying Li
- Academician Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China; Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Shuang Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nano-safety, Institute of High Energy Physics, Beijing 100049, China; CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Houxiang Hu
- Academician Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China; Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China.
| | - Zhanjun Gu
- Academician Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nano-safety, Institute of High Energy Physics, Beijing 100049, China; Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
7
|
Grandjean CE, Pedersen SF, Christensen C, Dibenedetto A, Eriksen T, Binderup T, Kjaer A. Imaging of atherosclerosis with [ 64Cu]Cu-DOTA-TATE in a translational head-to-head comparison study with [ 18F]FDG, and Na[ 18F]F in rabbits. Sci Rep 2023; 13:9249. [PMID: 37286582 DOI: 10.1038/s41598-023-35302-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 05/16/2023] [Indexed: 06/09/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease of the larger arteries that may lead to cardiovascular events. Identification of patients at highest risk of cardiovascular events is challenging, but molecular imaging using positron emission tomography (PET) may prove useful. The aim of this study was to evaluate and compare head-to-head three different PET tracers. Furthermore, tracer uptake is compared to gene expression alterations of the arterial vessel wall. Male New Zealand White rabbits (control group; n = 10, atherosclerotic group; n = 11) were used for the study. Vessel wall uptake was assessed with the three different PET tracers: [18F]FDG (inflammation), Na[18F]F (microcalcification), and [64Cu]Cu-DOTA-TATE (macrophages), using PET/computed tomography (CT). Tracer uptake was measured as standardized uptake value (SUV), and arteries from both groups were analyzed ex vivo by autoradiography, qPCR, histology, and immunohistochemistry. In rabbits, the atherosclerotic group showed significantly higher uptake of all three tracers compared to the control group [18F]FDG: SUVmean 1.50 ± 0.11 versus 1.23 ± 0.09, p = 0.025; Na[18F]F: SUVmean 1.54 ± 0.06 versus 1.18 ± 0.10, p = 0.006; and [64Cu]Cu-DOTA-TATE: SUVmean 2.30 ± 0.27 versus 1.65 ± 0.16; p = 0.047. Of the 102 genes analyzed, 52 were differentially expressed in the atherosclerotic group compared to the control group and several genes correlated with tracer uptake. In conclusion, we demonstrated the diagnostic value of [64Cu]Cu-DOTA-TATE and Na[18F]F for identifying atherosclerosis in rabbits. The two PET tracers provided information distinct from that obtained with [18F]FDG. None of the three tracers correlated significantly to each other, but [64Cu]Cu-DOTA-TATE and Na[18F]F uptake both correlated with markers of inflammation. [64Cu]Cu-DOTA-TATE was higher in atherosclerotic rabbits compared to [18F]FDG and Na[18F]F.
Collapse
Affiliation(s)
- Constance E Grandjean
- Department of Clinical Physiology, Nuclear Medicine and PET & Cluster for Molecular Imaging, Copenhagen University Hospital-Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Sune F Pedersen
- Department of Clinical Physiology, Nuclear Medicine and PET & Cluster for Molecular Imaging, Copenhagen University Hospital-Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Camilla Christensen
- Department of Clinical Physiology, Nuclear Medicine and PET & Cluster for Molecular Imaging, Copenhagen University Hospital-Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Altea Dibenedetto
- Department of Clinical Physiology, Nuclear Medicine and PET & Cluster for Molecular Imaging, Copenhagen University Hospital-Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Thomas Eriksen
- Department of Veterinary Clinical and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tina Binderup
- Department of Clinical Physiology, Nuclear Medicine and PET & Cluster for Molecular Imaging, Copenhagen University Hospital-Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine and PET & Cluster for Molecular Imaging, Copenhagen University Hospital-Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark.
| |
Collapse
|
8
|
Hsu JC, Tang Z, Eremina OE, Sofias AM, Lammers T, Lovell JF, Zavaleta C, Cai W, Cormode DP. Nanomaterial-based contrast agents. NATURE REVIEWS. METHODS PRIMERS 2023; 3:30. [PMID: 38130699 PMCID: PMC10732545 DOI: 10.1038/s43586-023-00211-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/20/2023] [Indexed: 12/23/2023]
Abstract
Medical imaging, which empowers the detection of physiological and pathological processes within living subjects, has a vital role in both preclinical and clinical diagnostics. Contrast agents are often needed to accompany anatomical data with functional information or to provide phenotyping of the disease in question. Many newly emerging contrast agents are based on nanomaterials as their high payloads, unique physicochemical properties, improved sensitivity and multimodality capacity are highly desired for many advanced forms of bioimaging techniques and applications. Here, we review the developments in the field of nanomaterial-based contrast agents. We outline important nanomaterial design considerations and discuss the effect on their physicochemical attributes, contrast properties and biological behaviour. We also describe commonly used approaches for formulating, functionalizing and characterizing these nanomaterials. Key applications are highlighted by categorizing nanomaterials on the basis of their X-ray, magnetic, nuclear, optical and/or photoacoustic contrast properties. Finally, we offer our perspectives on current challenges and emerging research topics as well as expectations for future advancements in the field.
Collapse
Affiliation(s)
- Jessica C. Hsu
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - Zhongmin Tang
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - Olga E. Eremina
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Alexandros Marios Sofias
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Jonathan F. Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Cristina Zavaleta
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - David P. Cormode
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
9
|
Zan C, An J, Wu Z, Li S. Engineering molecular nanoprobes to target early atherosclerosis: Precise diagnostic tools and promising therapeutic carriers. Nanotheranostics 2023; 7:327-344. [PMID: 37064609 PMCID: PMC10093416 DOI: 10.7150/ntno.82654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/02/2023] [Indexed: 04/18/2023] Open
Abstract
Atherosclerosis, an inflammation-driven chronic blood vessel disease, is a major contributor to devastating cardiovascular events, bringing serious social and economic burdens. Currently, non-invasive diagnostic and therapeutic techniques in combination with novel nanosized materials as well as established molecular targets are under active investigation to develop integrated molecular imaging approaches, precisely visualizing and/or even effectively reversing early-stage plaques. Besides, mechanistic investigation in the past decades provides many potent candidates extensively involved in the initiation and progression of atherosclerosis. Recent hotly-studied imaging nanoprobes for detecting early plaques mainly including optical nanoprobes, photoacoustic nanoprobes, magnetic resonance nanoprobes, positron emission tomography nanoprobes, and other dual- and multi-modality imaging nanoprobes, have been proven to be surface functionalized with important molecular targets, which occupy tailored physical and biological properties for atherogenesis. Of note, these engineering nanoprobes provide long blood-pool residence and specific molecular targeting, which allows efficient recognition of early-stage atherosclerotic plaques and thereby function as a novel type of precise diagnostic tools as well as potential therapeutic carriers of anti-atherosclerosis drugs. There have been no available nanoprobes applied in the clinics so far, although many newly emerged nanoprobes, as exemplified by aggregation-induced emission nanoprobes and TiO2 nanoprobes, have been tested for cell lines in vitro and atherogenic animal models in vivo, achieving good experimental effects. Therefore, there is an urgent call to translate these preclinical results for nanoprobes into clinical trials. For this reason, this review aims to give an overview of currently investigated nanoprobes in the context of atherosclerosis, summarize relevant published studies showing applications of different kinds of formulated nanoprobes in early detection and reverse of plaques, discuss recent advances and some limitations thereof, and provide some insights into the development of the new generation of more precise and efficient molecular nanoprobes, with a critical property of specifically targeting early atherosclerosis.
Collapse
Affiliation(s)
- Chunfang Zan
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, China
| | - Jie An
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, China
| | - Zhifang Wu
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, China
- ✉ Corresponding authors: Prof. Zhifang Wu, E-mail: . Prof. Sijin Li, E-mail:
| | - Sijin Li
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, China
- ✉ Corresponding authors: Prof. Zhifang Wu, E-mail: . Prof. Sijin Li, E-mail:
| |
Collapse
|
10
|
Parry R, Majeed K, Pixley F, Hillis GS, Francis RJ, Schultz CJ. Unravelling the role of macrophages in cardiovascular inflammation through imaging: a state-of-the-art review. Eur Heart J Cardiovasc Imaging 2022; 23:e504-e525. [PMID: 35993316 PMCID: PMC9671294 DOI: 10.1093/ehjci/jeac167] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 07/31/2022] [Indexed: 11/13/2022] Open
Abstract
Cardiovascular disease remains the leading cause of death and disability for patients across the world. Our understanding of atherosclerosis as a primary cholesterol issue has diversified, with a significant dysregulated inflammatory component that largely remains untreated and continues to drive persistent cardiovascular risk. Macrophages are central to atherosclerotic inflammation, and they exist along a functional spectrum between pro-inflammatory and anti-inflammatory extremes. Recent clinical trials have demonstrated a reduction in major cardiovascular events with some, but not all, anti-inflammatory therapies. The recent addition of colchicine to societal guidelines for the prevention of recurrent cardiovascular events in high-risk patients with chronic coronary syndromes highlights the real-world utility of this class of therapies. A highly targeted approach to modification of interleukin-1-dependent pathways shows promise with several novel agents in development, although excessive immunosuppression and resulting serious infection have proven a barrier to implementation into clinical practice. Current risk stratification tools to identify high-risk patients for secondary prevention are either inadequately robust or prohibitively expensive and invasive. A non-invasive and relatively inexpensive method to identify patients who will benefit most from novel anti-inflammatory therapies is required, a role likely to be fulfilled by functional imaging methods. This review article outlines our current understanding of the inflammatory biology of atherosclerosis, upcoming therapies and recent landmark clinical trials, imaging modalities (both invasive and non-invasive) and the current landscape surrounding functional imaging including through targeted nuclear and nanobody tracer development and their application.
Collapse
Affiliation(s)
- Reece Parry
- School of Medicine, University of Western Australia, Perth 6009, Australia
- Department of Cardiology, Royal Perth Hospital, 197 Wellington Street, Perth, WA 6000, Australia
| | - Kamran Majeed
- School of Medicine, University of Western Australia, Perth 6009, Australia
- Department of Cardiology, Waikato District Health Board, Hamilton 3204, New Zealand
| | - Fiona Pixley
- School of Biomedical Sciences, Pharmacology and Toxicology, University of Western Australia, Perth 6009, Australia
| | - Graham Scott Hillis
- School of Medicine, University of Western Australia, Perth 6009, Australia
- Department of Cardiology, Royal Perth Hospital, 197 Wellington Street, Perth, WA 6000, Australia
| | - Roslyn Jane Francis
- School of Medicine, University of Western Australia, Perth 6009, Australia
- Department of Nuclear Medicine, Sir Charles Gairdner Hospital, Perth 6009, Australia
| | - Carl Johann Schultz
- School of Medicine, University of Western Australia, Perth 6009, Australia
- Department of Cardiology, Royal Perth Hospital, 197 Wellington Street, Perth, WA 6000, Australia
| |
Collapse
|
11
|
Aranda-Lara L, Isaac-Olivé K, Ocampo-García B, Ferro-Flores G, González-Romero C, Mercado-López A, García-Marín R, Santos-Cuevas C, Estrada JA, Morales-Avila E. Engineered rHDL Nanoparticles as a Suitable Platform for Theranostic Applications. Molecules 2022; 27:7046. [PMID: 36296638 PMCID: PMC9610567 DOI: 10.3390/molecules27207046] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/12/2022] [Accepted: 10/14/2022] [Indexed: 08/27/2023] Open
Abstract
Reconstituted high-density lipoproteins (rHDLs) can transport and specifically release drugs and imaging agents, mediated by the Scavenger Receptor Type B1 (SR-B1) present in a wide variety of tumor cells, providing convenient platforms for developing theranostic systems. Usually, phospholipids or Apo-A1 lipoproteins on the particle surfaces are the motifs used to conjugate molecules for the multifunctional purposes of the rHDL nanoparticles. Cholesterol has been less addressed as a region to bind molecules or functional groups to the rHDL surface. To maximize the efficacy and improve the radiolabeling of rHDL theranostic systems, we synthesized compounds with bifunctional agents covalently linked to cholesterol. This strategy means that the radionuclide was bound to the surface, while the therapeutic agent was encapsulated in the lipophilic core. In this research, HYNIC-S-(CH2)3-S-Cholesterol and DOTA-benzene-p-SC-NH-(CH2)2-NH-Cholesterol derivatives were synthesized to prepare nanoparticles (NPs) of HYNIC-rHDL and DOTA-rHDL, which can subsequently be linked to radionuclides for SPECT/PET imaging or targeted radiotherapy. HYNIC is used to complexing 99mTc and DOTA for labeling molecules with 111, 113mIn, 67, 68Ga, 177Lu, 161Tb, 225Ac, and 64Cu, among others. In vitro studies showed that the NPs of HYNIC-rHDL and DOTA-rHDL maintain specific recognition by SR-B1 and the ability to internalize and release, in the cytosol of cancer cells, the molecules carried in their core. The biodistribution in mice showed a similar behavior between rHDL (without surface modification) and HYNIC-rHDL, while DOTA-rHDL exhibited a different biodistribution pattern due to the significant reduction in the lipophilicity of the modified cholesterol molecule. Both systems demonstrated characteristics for the development of suitable theranostic platforms for personalized cancer treatment.
Collapse
Affiliation(s)
- Liliana Aranda-Lara
- Faculty of Medicine, Universidad Autónoma del Estado de México, Toluca 50180, Estado de México, Mexico
| | - Keila Isaac-Olivé
- Faculty of Medicine, Universidad Autónoma del Estado de México, Toluca 50180, Estado de México, Mexico
| | - Blanca Ocampo-García
- Department of Radioactive Materials, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac 52750, Estado de México, Mexico
| | - Guillermina Ferro-Flores
- Department of Radioactive Materials, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac 52750, Estado de México, Mexico
| | - Carlos González-Romero
- Faculty of Chemistry, Universidad Autónoma del Estado de México, Toluca 50120, Estado de México, Mexico
| | - Alfredo Mercado-López
- Faculty of Chemistry, Universidad Autónoma del Estado de México, Toluca 50120, Estado de México, Mexico
| | - Rodrigo García-Marín
- Faculty of Chemistry, Universidad Autónoma del Estado de México, Toluca 50120, Estado de México, Mexico
| | - Clara Santos-Cuevas
- Department of Radioactive Materials, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac 52750, Estado de México, Mexico
| | - José A. Estrada
- Faculty of Medicine, Universidad Autónoma del Estado de México, Toluca 50180, Estado de México, Mexico
| | - Enrique Morales-Avila
- Faculty of Chemistry, Universidad Autónoma del Estado de México, Toluca 50120, Estado de México, Mexico
| |
Collapse
|
12
|
Li X, Wu M, Li J, Guo Q, Zhao Y, Zhang X. Advanced targeted nanomedicines for vulnerable atherosclerosis plaque imaging and their potential clinical implications. Front Pharmacol 2022; 13:906512. [PMID: 36313319 PMCID: PMC9606597 DOI: 10.3389/fphar.2022.906512] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 09/20/2022] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis plaques caused by cerebrovascular and coronary artery disease have been the leading cause of death and morbidity worldwide. Precise assessment of the degree of atherosclerotic plaque is critical for predicting the risk of atherosclerosis plaques and monitoring postinterventional outcomes. However, traditional imaging techniques to predict cardiocerebrovascular events mainly depend on quantifying the percentage reduction in luminal diameter, which would immensely underestimate non-stenotic high-risk plaque. Identifying the degree of atherosclerosis plaques still remains highly limited. vNanomedicine-based imaging techniques present unique advantages over conventional techniques due to the superior properties intrinsic to nanoscope, which possess enormous potential for characterization and detection of the features of atherosclerosis plaque vulnerability. Here, we review recent advancements in the development of targeted nanomedicine-based approaches and their applications to atherosclerosis plaque imaging and risk stratification. Finally, the challenges and opportunities regarding the future development and clinical translation of the targeted nanomedicine in related fields are discussed.
Collapse
Affiliation(s)
| | | | | | | | | | - Xuening Zhang
- Department of Radiology, Tianjin Medical University Second Hospital, Tianjin, China
| |
Collapse
|
13
|
Kondakov A, Berdalin A, Beregov M, Lelyuk V. Emerging Nuclear Medicine Imaging of Atherosclerotic Plaque Formation. J Imaging 2022; 8:261. [PMID: 36286355 PMCID: PMC9605050 DOI: 10.3390/jimaging8100261] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/01/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022] Open
Abstract
Atherosclerosis is a chronic widespread cardiovascular disease and a major predisposing factor for cardiovascular events, among which there are myocardial infarction and ischemic stroke. Atherosclerotic plaque formation is a process that involves different mechanisms, of which inflammation is the most common. Plenty of radiopharmaceuticals were developed to elucidate the process of plaque formation at different stages, some of which were highly specific for atherosclerotic plaque. This review summarizes the current nuclear medicine imaging landscape of preclinical and small-scale clinical studies of these specific RPs, which are not as widespread as labeled FDG, sodium fluoride, and choline. These include oxidation-specific epitope imaging, macrophage, and other cell receptors visualization, neoangiogenesis, and macrophage death imaging. It is shown that specific radiopharmaceuticals have strength in pathophysiologically sound imaging of the atherosclerotic plaques at different stages, but this also may induce problems with the signal registration for low-volume plaques in the vascular wall.
Collapse
Affiliation(s)
- Anton Kondakov
- Ultrasound and Functional Diagnostics Department, Federal Center of Brain Research and Neurotechnologies, 117513 Moscow, Russia
- Radiology and Radiotherapy Department, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Alexander Berdalin
- Ultrasound and Functional Diagnostics Department, Federal Center of Brain Research and Neurotechnologies, 117513 Moscow, Russia
| | - Mikhail Beregov
- Ultrasound and Functional Diagnostics Department, Federal Center of Brain Research and Neurotechnologies, 117513 Moscow, Russia
| | - Vladimir Lelyuk
- Ultrasound and Functional Diagnostics Department, Federal Center of Brain Research and Neurotechnologies, 117513 Moscow, Russia
| |
Collapse
|
14
|
Hu PP, Luo SX, Fan XQ, Li D, Tong XY. Macrophage-targeted nanomedicine for the diagnosis and management of atherosclerosis. Front Pharmacol 2022; 13:1000316. [PMID: 36160452 PMCID: PMC9501673 DOI: 10.3389/fphar.2022.1000316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 08/23/2022] [Indexed: 12/04/2022] Open
Abstract
Atherosclerosis is the primary cause of cardiovascular diseases, such as myocardial infarction and stroke, which account for the highest death toll worldwide. Macrophage is the major contributor to atherosclerosis progression, and therefore, macrophage-associated pathological process is considered an extremely important target for the diagnosis and treatment of atherosclerosis. However, the existing clinical strategies still have many bottlenecks and challenges in atherosclerosis’s early detection and management. Nanomedicine, using various nanoparticles/nanocarriers for medical purposes, can effectively load therapeutic agents, significantly improve their stability and accurately deliver them to the atherosclerotic plaques. In this review, we summarized the latest progress of the macrophage-targeted nanomedicine in the diagnosis and treatment of atherosclerosis, and their potential applications and clinical benefits are also discussed.
Collapse
Affiliation(s)
- Ping Ping Hu
- Chongqing Engineering Research Center for Pharmacodynamics Evaluation, College of Pharmacy, Chongqing Medical University, Chongqing, China
- *Correspondence: Ping Ping Hu, ; Xiao Yong Tong,
| | - Shuang Xue Luo
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Xiao Qing Fan
- Department of Thoracic Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Di Li
- Department of Pharmacy, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiao Yong Tong
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
- *Correspondence: Ping Ping Hu, ; Xiao Yong Tong,
| |
Collapse
|
15
|
Chen W, Schilperoort M, Cao Y, Shi J, Tabas I, Tao W. Macrophage-targeted nanomedicine for the diagnosis and treatment of atherosclerosis. Nat Rev Cardiol 2022; 19:228-249. [PMID: 34759324 PMCID: PMC8580169 DOI: 10.1038/s41569-021-00629-x] [Citation(s) in RCA: 199] [Impact Index Per Article: 99.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/22/2021] [Indexed: 12/12/2022]
Abstract
Nanotechnology could improve our understanding of the pathophysiology of atherosclerosis and contribute to the development of novel diagnostic and therapeutic strategies to further reduce the risk of cardiovascular disease. Macrophages have key roles in atherosclerosis progression and, therefore, macrophage-associated pathological processes are important targets for both diagnostic imaging and novel therapies for atherosclerosis. In this Review, we highlight efforts in the past two decades to develop imaging techniques and to therapeutically manipulate macrophages in atherosclerotic plaques with the use of rationally designed nanoparticles. We review the latest progress in nanoparticle-based imaging modalities that can specifically target macrophages. Using novel molecular imaging technology, these modalities enable the identification of advanced atherosclerotic plaques and the assessment of the therapeutic efficacy of medical interventions. Additionally, we provide novel perspectives on how macrophage-targeting nanoparticles can deliver a broad range of therapeutic payloads to atherosclerotic lesions. These nanoparticles can suppress pro-atherogenic macrophage processes, leading to improved resolution of inflammation and stabilization of plaques. Finally, we propose future opportunities for novel diagnostic and therapeutic strategies and provide solutions to challenges in this area for the purpose of accelerating the clinical translation of nanomedicine for the treatment of atherosclerotic vascular disease.
Collapse
Affiliation(s)
- Wei Chen
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Maaike Schilperoort
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Yihai Cao
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Ira Tabas
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA.
| | - Wei Tao
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
Hu Q, Fang Z, Ge J, Li H. Nanotechnology for cardiovascular diseases. Innovation (N Y) 2022; 3:100214. [PMID: 35243468 PMCID: PMC8866095 DOI: 10.1016/j.xinn.2022.100214] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/30/2022] [Accepted: 01/30/2022] [Indexed: 11/23/2022] Open
Abstract
Cardiovascular diseases have become the major killers in today's world, among which coronary artery diseases (CADs) make the greatest contributions to morbidity and mortality. Although state-of-the-art technologies have increased our knowledge of the cardiovascular system, the current diagnosis and treatment modalities for CADs still have limitations. As an emerging cross-disciplinary approach, nanotechnology has shown great potential for clinical use. In this review, recent advances in nanotechnology in the diagnosis of CADs will first be elucidated. Both the sensitivity and specificity of biosensors for biomarker detection and molecular imaging strategies, such as magnetic resonance imaging, optical imaging, nuclear scintigraphy, and multimodal imaging strategies, have been greatly increased with the assistance of nanomaterials. Second, various nanomaterials, such as liposomes, polymers (PLGA), inorganic nanoparticles (AuNPs, MnO2, etc.), natural nanoparticles (HDL, HA), and biomimetic nanoparticles (cell-membrane coating) will be discussed as engineered as drug (chemicals, proteins, peptides, and nucleic acids) carriers targeting pathological sites based on their optimal physicochemical properties and surface modification potential. Finally, some of these nanomaterials themselves are regarded as pharmaceuticals for the treatment of atherosclerosis because of their intrinsic antioxidative/anti-inflammatory and photoelectric/photothermal characteristics in a complex plaque microenvironment. In summary, novel nanotechnology-based research in the process of clinical transformation could continue to expand the horizon of nanoscale technologies in the diagnosis and therapy of CADs in the foreseeable future.
Collapse
Affiliation(s)
- Qinqin Hu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Shanghai Xuhui District Central Hospital & Zhongshan-xuhui Hospital, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Zheyan Fang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Shanghai Xuhui District Central Hospital & Zhongshan-xuhui Hospital, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Shanghai Xuhui District Central Hospital & Zhongshan-xuhui Hospital, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Hua Li
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Shanghai Xuhui District Central Hospital & Zhongshan-xuhui Hospital, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
17
|
Pan Q, Xu J, Wen CJ, Xiong YY, Gong ZT, Yang YJ. Nanoparticles: Promising Tools for the Treatment and Prevention of Myocardial Infarction. Int J Nanomedicine 2021; 16:6719-6747. [PMID: 34621124 PMCID: PMC8491866 DOI: 10.2147/ijn.s328723] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/17/2021] [Indexed: 12/12/2022] Open
Abstract
Despite several recent advances, current therapy and prevention strategies for myocardial infarction are far from satisfactory, owing to limitations in their applicability and treatment effects. Nanoparticles (NPs) enable the targeted and stable delivery of therapeutic compounds, enhance tissue engineering processes, and regulate the behaviour of transplants such as stem cells. Thus, NPs may be more effective than other mechanisms, and may minimize potential adverse effects. This review provides evidence for the view that function-oriented systems are more practical than traditional material-based systems; it also summarizes the latest advances in NP-based strategies for the treatment and prevention of myocardial infarction.
Collapse
Affiliation(s)
- Qi Pan
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Jing Xu
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Cen-Jin Wen
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Yu-Yan Xiong
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Zhao-Ting Gong
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Yue-Jin Yang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| |
Collapse
|
18
|
Van Valkenburgh J, Meuret C, Martinez AE, Kodancha V, Solomon V, Chen K, Yassine HN. Understanding the Exchange of Systemic HDL Particles Into the Brain and Vascular Cells Has Diagnostic and Therapeutic Implications for Neurodegenerative Diseases. Front Physiol 2021; 12:700847. [PMID: 34552500 PMCID: PMC8450374 DOI: 10.3389/fphys.2021.700847] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/29/2021] [Indexed: 12/02/2022] Open
Abstract
High-density lipoproteins (HDLs) are complex, heterogenous lipoprotein particles, consisting of a large family of apolipoproteins, formed in subspecies of distinct shapes, sizes, and functions and are synthesized in both the brain and the periphery. HDL apolipoproteins are important determinants of Alzheimer’s disease (AD) pathology and vascular dementia, having both central and peripheral effects on brain amyloid-beta (Aβ) accumulation and vascular functions, however, the extent to which HDL particles (HLD-P) can exchange their protein and lipid components between the central nervous system (CNS) and the systemic circulation remains unclear. In this review, we delineate how HDL’s structure and composition enable exchange between the brain, cerebrospinal fluid (CSF) compartment, and vascular cells that ultimately affect brain amyloid metabolism and atherosclerosis. Accordingly, we then elucidate how modifications of HDL-P have diagnostic and therapeutic potential for brain vascular and neurodegenerative diseases.
Collapse
Affiliation(s)
- Juno Van Valkenburgh
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Cristiana Meuret
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Ashley E Martinez
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Vibha Kodancha
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Victoria Solomon
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Kai Chen
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Hussein N Yassine
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
19
|
Schrijver DP, Dreu A, Hofstraat SRJ, Kluza E, Zwolsman R, Deckers J, Anbergen T, Bruin K, Trines MM, Nugraha EG, Ummels F, Röring RJ, Beldman TJ, Teunissen AJP, Fayad ZA, Meel R, Mulder WJM. Nanoengineering Apolipoprotein A1‐Based Immunotherapeutics. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- David P. Schrijver
- Laboratory of Chemical Biology Department of Biomedical Engineering Eindhoven University of Technology Eindhoven 5612 AZ The Netherlands
| | - Anne Dreu
- Laboratory of Chemical Biology Department of Biomedical Engineering Eindhoven University of Technology Eindhoven 5612 AZ The Netherlands
| | - Stijn R. J. Hofstraat
- Laboratory of Chemical Biology Department of Biomedical Engineering Eindhoven University of Technology Eindhoven 5612 AZ The Netherlands
| | - Ewelina Kluza
- Laboratory of Chemical Biology Department of Biomedical Engineering Eindhoven University of Technology Eindhoven 5612 AZ The Netherlands
| | - Robby Zwolsman
- Laboratory of Chemical Biology Department of Biomedical Engineering Eindhoven University of Technology Eindhoven 5612 AZ The Netherlands
| | - Jeroen Deckers
- Laboratory of Chemical Biology Department of Biomedical Engineering Eindhoven University of Technology Eindhoven 5612 AZ The Netherlands
| | - Tom Anbergen
- Laboratory of Chemical Biology Department of Biomedical Engineering Eindhoven University of Technology Eindhoven 5612 AZ The Netherlands
| | - Koen Bruin
- Laboratory of Chemical Biology Department of Biomedical Engineering Eindhoven University of Technology Eindhoven 5612 AZ The Netherlands
| | - Mirre M. Trines
- Laboratory of Chemical Biology Department of Biomedical Engineering Eindhoven University of Technology Eindhoven 5612 AZ The Netherlands
| | - Eveline G. Nugraha
- Laboratory of Chemical Biology Department of Biomedical Engineering Eindhoven University of Technology Eindhoven 5612 AZ The Netherlands
| | - Floor Ummels
- Laboratory of Chemical Biology Department of Biomedical Engineering Eindhoven University of Technology Eindhoven 5612 AZ The Netherlands
| | - Rutger J. Röring
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI) Radboud University Nijmegen Medical Center Nijmegen 6525 GA The Netherlands
| | - Thijs J. Beldman
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI) Radboud University Nijmegen Medical Center Nijmegen 6525 GA The Netherlands
| | - Abraham J. P. Teunissen
- Biomedical Engineering and Imaging Institute Icahn School of Medicine at Mount Sinai New York NY 10029‐6574 USA
| | - Zahi A. Fayad
- Biomedical Engineering and Imaging Institute Icahn School of Medicine at Mount Sinai New York NY 10029‐6574 USA
| | - Roy Meel
- Laboratory of Chemical Biology Department of Biomedical Engineering Eindhoven University of Technology Eindhoven 5612 AZ The Netherlands
| | - Willem J. M. Mulder
- Laboratory of Chemical Biology Department of Biomedical Engineering Eindhoven University of Technology Eindhoven 5612 AZ The Netherlands
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI) Radboud University Nijmegen Medical Center Nijmegen 6525 GA The Netherlands
| |
Collapse
|
20
|
Li Z, Tang H, Tu Y. Molecular and Nonmolecular Imaging of Macrophages in Atherosclerosis. Front Cardiovasc Med 2021; 8:670639. [PMID: 34095259 PMCID: PMC8169961 DOI: 10.3389/fcvm.2021.670639] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 04/22/2021] [Indexed: 12/28/2022] Open
Abstract
Atherosclerosis is a major cause of ischemic heart disease, and the increasing medical burden associated with atherosclerotic cardiovascular disease has become a major public health concern worldwide. Macrophages play an important role in all stages of the dynamic progress of atherosclerosis, from its initiation and lesion expansion increasing the vulnerability of plaques, to the formation of unstable plaques and clinical manifestations. Early imaging can identify patients at risk of coronary atherosclerotic disease and its complications, enabling preventive measures to be initiated. Recent advances in molecular imaging have involved the noninvasive and semi-quantitative targeted imaging of macrophages and their related molecules in vivo, which can detect atheroma earlier and more accurately than conventional imaging. Multimodal imaging integrates vascular structure, function, and molecular imaging technology to achieve multi-dimensional imaging, which can be used to comprehensively evaluate blood vessels and obtain clinical information based on anatomical structure and molecular level. At the same time, the rapid development of nonmolecular imaging technologies, such as intravascular imaging, which have the unique advantages of having intuitive accuracy and providing rich information to identify macrophage inflammation and inform targeted personalized treatment, has also been seen. In this review, we highlight recent methods and research hotspots in molecular and nonmolecular imaging of macrophages in atherosclerosis that have enormous potential for rapid clinical application.
Collapse
Affiliation(s)
- Zhaoyue Li
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hao Tang
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yingfeng Tu
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
21
|
B Uribe K, Benito-Vicente A, Martin C, Blanco-Vaca F, Rotllan N. (r)HDL in theranostics: how do we apply HDL's biology for precision medicine in atherosclerosis management? Biomater Sci 2021; 9:3185-3208. [PMID: 33949389 DOI: 10.1039/d0bm01838d] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
High-density lipoproteins (HDL) are key players in cholesterol metabolism homeostasis since they are responsible for transporting excess cholesterol from peripheral tissues to the liver. Imbalance in this process, due to either excessive accumulation or impaired clearance, results in net cholesterol accumulation and increases the risk of cardiovascular disease (CVD). Therefore, significant effort has been focused on the development of therapeutic tools capable of either directly or indirectly enhancing HDL-guided reverse cholesterol transport (RCT). More recently, in light of the emergence of precision nanomedicine, there has been renewed research interest in attempting to take advantage of the development of advanced recombinant HDL (rHDL) for both therapeutic and diagnostic purposes. In this review, we provide an update on the different approaches that have been developed using rHDL, focusing on the rHDL production methodology and rHDL applications in theranostics. We also compile a series of examples highlighting potential future perspectives in the field.
Collapse
Affiliation(s)
- Kepa B Uribe
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramon 182, 20014, Donostia San Sebastián, Spain.
| | - Asier Benito-Vicente
- Instituto Biofisika (UPV/EHU, CSIC) and Departamento de Bioquímica, Universidad del País Vasco, Apdo.644, 48080 Bilbao, Spain.
| | - Cesar Martin
- Instituto Biofisika (UPV/EHU, CSIC) and Departamento de Bioquímica, Universidad del País Vasco, Apdo.644, 48080 Bilbao, Spain.
| | - Francisco Blanco-Vaca
- Servei de Bioquímica, Hospital Santa Creu i Sant Pau-Institut d'Investigacions Biomèdiques (IIB) Sant Pau, 08041 Barcelona, Spain. and CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain and Departament de Bioquímica i Biología Molecular, Universitat Autònoma de Barcelona, Spain and Institut de Recerca de l'Hospital Santa Creu i Sant Pau-Institut d'Investigacions Biomèdiques (IIB) Sant Pau, 08025 Barcelona, Spain.
| | - Noemi Rotllan
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain and Institut de Recerca de l'Hospital Santa Creu i Sant Pau-Institut d'Investigacions Biomèdiques (IIB) Sant Pau, 08025 Barcelona, Spain.
| |
Collapse
|
22
|
Sobol NB, Korsen JA, Younes A, Edwards KJ, Lewis JS. ImmunoPET Imaging of Pancreatic Tumors with 89Zr-Labeled Gold Nanoparticle-Antibody Conjugates. Mol Imaging Biol 2021; 23:84-94. [PMID: 32909244 PMCID: PMC7785666 DOI: 10.1007/s11307-020-01535-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/30/2020] [Accepted: 08/18/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE Targeted delivery in vivo remains an immense roadblock for the translation of nanomaterials into the clinic. The greatest obstacle is the mononuclear phagocyte system (MPS), which sequesters foreign substances from general circulation and causes accumulation in organs such as the liver and spleen. The purpose of this study was to determine whether attaching an active targeting antibody, 5B1, to the surface of gold nanoparticles and using clodronate liposomes to deplete liver and splenic macrophages could help to minimize uptake by MPS organs, increase targeted delivery to CA19.9-positive pancreatic tumors, and enhance pancreatic tumor delineation. PROCEDURES To produce the antibody-gold nanoparticle conjugate (Ab-AuNP), the Ab was conjugated to p-isothiocyanatobenzyl-desferrioxamine (p-SCN-DFO) and subsequently conjugated to NHS-activated gold nanoparticles. The Ab-AuNP was characterized by transmission electron microscopy (TEM) and atomic force microscopy (AFM). Modified Lindmo assay was performed to assess binding affinity and internalization potential in vitro. The Ab-AuNP was radiolabeled with 89Zr and injected into CA19.9-positive BxPc-3 pancreatic orthotopic tumor-bearing mice pretreated with or without clodronate liposomes for PET imaging and biodistribution studies. Inductively coupled plasma-optical emission spectrometry (ICP-OES) analysis was used to confirm delivery of gold nanoparticles to BxPc-3 pancreatic subcutaneous xenografts. RESULTS Mice pretreated with clodronate liposomes in an orthotopic setting demonstrated decreased liver uptake at early time points (12.2 ± 2.3 % ID/g vs. 22.8 ± 3.8 % ID/g at 24 h) and increased tumor uptake at 120 h (13.8 ± 8.0 % ID/g vs. 6.0 ± 1.2 % ID/g). This allowed for delineation of orthotopic pancreatic xenografts in significantly more mice treated with clodronate (6/6) than in mice not treated with clodronate (2/6) or mice injected with gold nanoparticles labeled with a nonspecific antibody (0/5). CONCLUSIONS The combination of clodronate liposomes and an active targeting antibody on the surface of gold nanoparticles allowed for PET/CT imaging of subcutaneous and orthotopic pancreatic xenografts in mice.
Collapse
Affiliation(s)
- Nicholas B Sobol
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joshua A Korsen
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pharmacology, Weill Cornell Medical College, New York, NY, USA
| | - Ali Younes
- Department of Chemistry, Hunter College, New York, NY, USA
| | - Kimberly J Edwards
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Pharmacology, Weill Cornell Medical College, New York, NY, USA.
- Program in Molecular Pharmacology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
23
|
Osborn EA, Albaghdadi M, Libby P, Jaffer FA. Molecular Imaging of Atherosclerosis. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00086-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
24
|
Pedersbæk D, Simonsen JB. A systematic review of the biodistribution of biomimetic high-density lipoproteins in mice. J Control Release 2020; 328:792-804. [PMID: 32971201 DOI: 10.1016/j.jconrel.2020.09.038] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/18/2022]
Abstract
For the past two decades, biomimetic high-density lipoproteins (b-HDL) have been used for various drug delivery applications. The b-HDL mimic the endogenous HDL, and therefore possess many attractive features for drug delivery, including high biocompatibility, biodegradability, and ability to transport and deliver their cargo (e.g. drugs and/or imaging agents) to specific cells and tissues that are recognized by HDL. The b-HDL designs reported in the literature often differ in size, shape, composition, and type of incorporated cargo. However, there exists only limited insight into how the b-HDL design dictates their biodistribution. To fill this gap, we conducted a comprehensive systematic literature search of biodistribution studies using various designs of apolipoprotein A-I (apoA-I)-based b-HDL (i.e. b-HDL with apoA-I, apoA-I mutants, or apoA-I mimicking peptides). We carefully screened 679 papers (search hits) for b-HDL biodistribution studies in mice, and ended up with 24 relevant biodistribution profiles that we compared according to b-HDL design. We show similarities between b-HDL biodistribution studies irrespectively of the b-HDL design, whereas the biodistribution of the b-HDL components (lipids and scaffold) differ significantly. The b-HDL lipids primarily accumulate in liver, while the b-HDL scaffold primarily accumulates in the kidney. Furthermore, both b-HDL lipids and scaffold accumulate well in the tumor tissue in tumor-bearing mice. Finally, we present essential considerations and strategies for b-HDL labeling, and discuss how the b-HDL biodistribution can be tuned through particle design and administration route. Our meta-analysis and discussions provide a detailed overview of the fate of b-HDL in mice that is highly relevant when applying b-HDL for drug delivery or in vivo imaging applications.
Collapse
Affiliation(s)
- Dennis Pedersbæk
- Technical University of Denmark, Department of Health Technology, 2800 Kgs. Lyngby, Denmark
| | - Jens B Simonsen
- Technical University of Denmark, Department of Health Technology, 2800 Kgs. Lyngby, Denmark.
| |
Collapse
|
25
|
Assessment of Microvessel Permeability in Murine Atherosclerotic Vein Grafts Using Two-Photon Intravital Microscopy. Int J Mol Sci 2020; 21:ijms21239244. [PMID: 33287463 PMCID: PMC7730593 DOI: 10.3390/ijms21239244] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 11/26/2020] [Accepted: 12/01/2020] [Indexed: 01/25/2023] Open
Abstract
Plaque angiogenesis and plaque hemorrhage are major players in the destabilization and rupture of atherosclerotic lesions. As these are dynamic processes, imaging of plaque angiogenesis, especially the integrity or leakiness of angiogenic vessels, can be an extremely useful tool in the studies on atherosclerosis pathophysiology. Visualizing plaque microvessels in 3D would enable us to study the architecture and permeability of adventitial and intimal plaque microvessels in advanced atherosclerotic lesions. We hypothesized that a comparison of the vascular permeability between healthy continuous and fenestrated as well as diseased leaky microvessels, would allow us to evaluate plaque microvessel leakiness. We developed and validated a two photon intravital microscopy (2P-IVM) method to assess the leakiness of plaque microvessels in murine atherosclerosis-prone ApoE3*Leiden vein grafts based on the quantification of fluorescent-dextrans extravasation in real-time. We describe a novel 2P-IVM set up to study vessels in the neck region of living mice. We show that microvessels in vein graft lesions are in their pathological state more permeable in comparison with healthy continuous and fenestrated microvessels. This 2P-IVM method is a promising approach to assess plaque angiogenesis and leakiness. Moreover, this method is an important advancement to validate therapeutic angiogenic interventions in preclinical atherosclerosis models.
Collapse
|
26
|
Modak M, Frey MA, Yi S, Liu Y, Scott EA. Employment of targeted nanoparticles for imaging of cellular processes in cardiovascular disease. Curr Opin Biotechnol 2020; 66:59-68. [PMID: 32682272 PMCID: PMC7744313 DOI: 10.1016/j.copbio.2020.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/13/2020] [Accepted: 06/07/2020] [Indexed: 12/18/2022]
Abstract
Cardiovascular disease (CVD) is a leading cause of global mortality, accounting for pathologies that are primarily of atherosclerotic origin and driven by specific cell populations. A need exists for effective, non-invasive methods to assess the risk of potentially fatal major adverse cardiovascular events (MACE) before occurrence and to monitor post-interventional outcomes such as tissue regeneration. Molecular imaging has widespread applications in CVD diagnostic assessment, through modalities including magnetic resonance imaging (MRI), positron emission tomography (PET), and acoustic imaging methods. However, current gold-standard small molecule contrast agents are not cell-specific, relying on non-specific uptake to facilitate imaging of biologic processes. Nanomaterials can be engineered for targeted delivery to specific cell populations, and several nanomaterial systems have been developed for pre-clinical molecular imaging. Here, we review recent advances in nanoparticle-mediated approaches for imaging of cellular processes in cardiovascular disease, focusing on efforts to detect inflammation, assess lipid accumulation, and monitor tissue regeneration.
Collapse
Affiliation(s)
- Mallika Modak
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | - Molly A Frey
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL, USA; Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA
| | - Sijia Yi
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL, USA
| | - Yugang Liu
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL, USA
| | - Evan A Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA; Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL, USA; Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA; Department of Microbiology-Immunology, Northwestern University, Chicago, IL 60611, USA; Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
27
|
Surmounting the endothelial barrier for delivery of drugs and imaging tracers. Atherosclerosis 2020; 315:93-101. [DOI: 10.1016/j.atherosclerosis.2020.04.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/14/2020] [Accepted: 04/29/2020] [Indexed: 12/18/2022]
|
28
|
Fadeel B, Alexiou C. Brave new world revisited: Focus on nanomedicine. Biochem Biophys Res Commun 2020; 533:36-49. [PMID: 32921412 DOI: 10.1016/j.bbrc.2020.08.046] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 12/11/2022]
Abstract
Nanomedicine is at a crossroads: with relatively few success stories in terms of clinical translation despite more and more research on increasingly sophisticated nanomaterials, it is important to consider whether we are on the right track. Indeed, it is crucial that we address the fact that while considerable efforts are being made to overcome barriers to translation from the bench to the clinic, scientists are still struggling to decipher fundamental aspects of nanomaterial interactions with biological systems. We believe that a key to the successful adoption of nanomedicines in oncology and beyond lies in a deeper understanding of underlying biological processes and in decoding interactions between engineered nanomaterials and biological systems. Here we provide an overview of progress in nanomedicine during the past 5 years.
Collapse
Affiliation(s)
- Bengt Fadeel
- Nanomedicine & Nanosafety Laboratory (NNL), Division of Molecular Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77, Stockholm, Sweden.
| | - Christoph Alexiou
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Section for Experimental Oncology and Nanomedicine (SEON), University Hospital Erlangen, Else Kröner-Fresenius-Stiftung Professorship, 91054, Erlangen, Germany
| |
Collapse
|
29
|
Ćorović A, Wall C, Mason JC, Rudd JHF, Tarkin JM. Novel Positron Emission Tomography Tracers for Imaging Vascular Inflammation. Curr Cardiol Rep 2020; 22:119. [PMID: 32772188 PMCID: PMC7415747 DOI: 10.1007/s11886-020-01372-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Purpose of Review To provide a focused update on recent advances in positron emission tomography (PET) imaging in vascular inflammatory diseases and consider future directions in the field. Recent Findings While PET imaging with 18F-fluorodeoxyglucose (FDG) can provide a useful marker of disease activity in several vascular inflammatory diseases, including atherosclerosis and large-vessel vasculitis, this tracer lacks inflammatory cell specificity and is not a practical solution for imaging the coronary vasculature because of avid background myocardial signal. To overcome these limitations, research is ongoing to identify novel PET tracers that can more accurately track individual components of vascular immune responses. Use of these novel PET tracers could lead to a better understanding of underlying disease mechanisms and help inform the identification and stratification of patients for newly emerging immune-modulatory therapies. Summary Future research is needed to realise the true clinical translational value of PET imaging in vascular inflammatory diseases.
Collapse
Affiliation(s)
- Andrej Ćorović
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Christopher Wall
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Justin C Mason
- Cardiovascular Division, National Heart & Lung Institute, Imperial College London, London, UK
| | - James H F Rudd
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Jason M Tarkin
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK. .,Cardiovascular Division, National Heart & Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
30
|
Georgilis E, Abdelghani M, Pille J, Aydinlioglu E, van Hest JC, Lecommandoux S, Garanger E. Nanoparticles based on natural, engineered or synthetic proteins and polypeptides for drug delivery applications. Int J Pharm 2020; 586:119537. [DOI: 10.1016/j.ijpharm.2020.119537] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/03/2020] [Accepted: 06/06/2020] [Indexed: 12/12/2022]
|
31
|
Yoon JK, Park BN, Ryu EK, An YS, Lee SJ. Current Perspectives on 89Zr-PET Imaging. Int J Mol Sci 2020; 21:ijms21124309. [PMID: 32560337 PMCID: PMC7352467 DOI: 10.3390/ijms21124309] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/13/2020] [Accepted: 06/15/2020] [Indexed: 12/14/2022] Open
Abstract
89Zr is an emerging radionuclide that plays an essential role in immuno-positron emission tomography (PET) imaging. The long half-life of 89Zr (t1/2 = 3.3 days) is favorable for evaluating the in vivo distribution of monoclonal antibodies. Thus, the use of 89Zr is promising for monitoring antibody-based cancer therapies. Immuno-PET combines the sensitivity of PET with the specificity of antibodies. A number of studies have been conducted to investigate the feasibility of 89Zr immuno-PET imaging for predicting the efficacy of radioimmunotherapy and antibody therapies, imaging target expression, detecting target-expressing tumors, and the monitoring of anti-cancer chemotherapies. In this review, we summarize the current status of PET imaging using 89Zr in both preclinical and clinical studies by highlighting the use of immuno-PET for the targets of high clinical relevance. We also present 89Zr-PET applications other than immuno-PET, such as nanoparticle imaging and cell tracking. Finally, we discuss the limitations and the ongoing research being performed to overcome the remaining hurdles.
Collapse
Affiliation(s)
- Joon-Kee Yoon
- Department of Nuclear Medicine & Molecular Imaging, Ajou University School of Medicine, Worldcup-ro 164, Suwon 16499, Korea; (B.-N.P.); (Y.-S.A.); (S.-J.L.)
- Correspondence: ; Tel.: +82-31-219-4303
| | - Bok-Nam Park
- Department of Nuclear Medicine & Molecular Imaging, Ajou University School of Medicine, Worldcup-ro 164, Suwon 16499, Korea; (B.-N.P.); (Y.-S.A.); (S.-J.L.)
| | - Eun-Kyoung Ryu
- Division of Magnetic Resonance, Korea Basic Science Institute, 162, Yeongudanji-ro, Cheongju 28119, Korea;
| | - Young-Sil An
- Department of Nuclear Medicine & Molecular Imaging, Ajou University School of Medicine, Worldcup-ro 164, Suwon 16499, Korea; (B.-N.P.); (Y.-S.A.); (S.-J.L.)
| | - Su-Jin Lee
- Department of Nuclear Medicine & Molecular Imaging, Ajou University School of Medicine, Worldcup-ro 164, Suwon 16499, Korea; (B.-N.P.); (Y.-S.A.); (S.-J.L.)
| |
Collapse
|
32
|
Radiolabeled liposomes and lipoproteins as lipidic nanoparticles for imaging and therapy. Chem Phys Lipids 2020; 230:104934. [PMID: 32562666 DOI: 10.1016/j.chemphyslip.2020.104934] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/09/2020] [Accepted: 06/15/2020] [Indexed: 02/06/2023]
Abstract
Radiolabeled lipidic nanoparticles, particularly liposomes and lipoproteins, are of great interest as agents for imaging and therapy, due not only to their peculiar physicochemical and biological properties, but also to their great versatility and the ability to manipulate them to obtain the desired properties. This review provides an overview of radionuclide labeling strategies for preparing diagnostic and therapeutic nanoparticles based on liposomes and lipoproteins that have been developed to date, as well as the main quality control methods and in vivo applications.
Collapse
|
33
|
Li X, Rosenkrans ZT, Wang J, Cai W. PET imaging of macrophages in cardiovascular diseases. Am J Transl Res 2020; 12:1491-1514. [PMID: 32509158 PMCID: PMC7270023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 03/14/2020] [Indexed: 06/11/2023]
Abstract
Cardiovascular diseases (CVDs) have been the leading cause of death in United States. While tremendous progress has been made for treating CVDs over the year, the high prevalence and substantial medical costs requires the necessity for novel methods for the early diagnosis and treatment monitoring of CVDs. Macrophages are a promising target due to its crucial role in the progress of CVDs (atherosclerosis, myocardial infarction and inflammatory cardiomyopathies). Positron emission tomography (PET) is a noninvasive imaging technique with high sensitivity and provides quantitive functional information of the macrophages in CVDs. Although 18F-FDG can be taken up by active macrophages, the PET imaging tracer is non-specific and susceptible to blood glucose levels. Thus, developing more specific PET tracers will help us understand the role of macrophages in CVDs. Moreover, macrophage-targeted PET imaging will further improve the diagnosis, treatment monitoring, and outcome prediction for patients with CVDs. In this review, we summarize various targets-based tracers for the PET imaging of macrophages in CVDs and highlight research gaps to advise future directions.
Collapse
Affiliation(s)
- Xiang Li
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical UniversityXi’an 710032, Shaanxi, China
- Department of Radiology and Medical Physics, University of Wisconsin-MadisonMadison, WI 53705, USA
| | - Zachary T Rosenkrans
- Department of Pharmaceutical Sciences, University of Wisconsin-MadisonMadison, WI 53705, USA
| | - Jing Wang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical UniversityXi’an 710032, Shaanxi, China
| | - Weibo Cai
- Department of Radiology and Medical Physics, University of Wisconsin-MadisonMadison, WI 53705, USA
- Department of Pharmaceutical Sciences, University of Wisconsin-MadisonMadison, WI 53705, USA
| |
Collapse
|
34
|
Senders ML, Meerwaldt AE, van Leent MMT, Sanchez-Gaytan BL, van de Voort JC, Toner YC, Maier A, Klein ED, Sullivan NAT, Sofias AM, Groenen H, Faries C, Oosterwijk RS, van Leeuwen EM, Fay F, Chepurko E, Reiner T, Duivenvoorden R, Zangi L, Dijkhuizen RM, Hak S, Swirski FK, Nahrendorf M, Pérez-Medina C, Teunissen AJP, Fayad ZA, Calcagno C, Strijkers GJ, Mulder WJM. Probing myeloid cell dynamics in ischaemic heart disease by nanotracer hot-spot imaging. NATURE NANOTECHNOLOGY 2020; 15:398-405. [PMID: 32313216 PMCID: PMC7416336 DOI: 10.1038/s41565-020-0642-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 01/16/2020] [Indexed: 05/19/2023]
Abstract
Ischaemic heart disease evokes a complex immune response. However, tools to track the systemic behaviour and dynamics of leukocytes non-invasively in vivo are lacking. Here, we present a multimodal hot-spot imaging approach using an innovative high-density lipoprotein-derived nanotracer with a perfluoro-crown ether payload (19F-HDL) to allow myeloid cell tracking by 19F magnetic resonance imaging. The 19F-HDL nanotracer can additionally be labelled with zirconium-89 and fluorophores to detect myeloid cells by in vivo positron emission tomography imaging and optical modalities, respectively. Using our nanotracer in atherosclerotic mice with myocardial infarction, we observed rapid myeloid cell egress from the spleen and bone marrow by in vivo 19F-HDL magnetic resonance imaging. Concurrently, using ex vivo techniques, we showed that circulating pro-inflammatory myeloid cells accumulated in atherosclerotic plaques and at the myocardial infarct site. Our multimodality imaging approach is a valuable addition to the immunology toolbox, enabling the study of complex myeloid cell behaviour dynamically.
Collapse
Affiliation(s)
- Max L Senders
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medical Biochemistry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Anu E Meerwaldt
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht/Utrecht University, Utrecht, the Netherlands
| | - Mandy M T van Leent
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medical Biochemistry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Brenda L Sanchez-Gaytan
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Instituto de Ciencias ICUAP, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Jan C van de Voort
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yohana C Toner
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexander Maier
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emma D Klein
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nathaniel A T Sullivan
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexandros Marios Sofias
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Hannah Groenen
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christopher Faries
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Roderick S Oosterwijk
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Esther M van Leeuwen
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Francois Fay
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Institut Galien Paris Sud, Faculté de Pharmacie, CNRS, Université Paris-Sud, Université Paris-Saclay, Paris, France
| | - Elena Chepurko
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY, USA
| | - Raphael Duivenvoorden
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Lior Zangi
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rick M Dijkhuizen
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht/Utrecht University, Utrecht, the Netherlands
| | - Sjoerd Hak
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Filip K Swirski
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Carlos Pérez-Medina
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Abraham J P Teunissen
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zahi A Fayad
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Claudia Calcagno
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gustav J Strijkers
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Willem J M Mulder
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Medical Biochemistry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
35
|
Calcagno C, Pérez-Medina C, Mulder WJM, Fayad ZA. Whole-Body Atherosclerosis Imaging by Positron Emission Tomography/Magnetic Resonance Imaging: From Mice to Nonhuman Primates. Arterioscler Thromb Vasc Biol 2020; 40:1123-1134. [PMID: 32237905 DOI: 10.1161/atvbaha.119.313629] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cardiovascular disease due to atherosclerosis is still the main cause of morbidity and mortality worldwide. This disease is a complex systemic disorder arising from a network of pathological processes within the arterial vessel wall, and, outside of the vasculature, in the hematopoietic system and organs involved in metabolism. Recent years have seen tremendous efforts in the development and validation of quantitative imaging technologies for the noninvasive evaluation of patients with atherosclerotic cardiovascular disease. Specifically, the advent of combined positron emission tomography and magnetic resonance imaging scanners has opened new exciting opportunities in cardiovascular imaging. In this review, we will describe how combined positron emission tomography/magnetic resonance imaging scanners can be leveraged to evaluate atherosclerotic cardiovascular disease at the whole-body level, with specific focus on preclinical animal models of disease, from mouse to nonhuman primates. We will broadly describe 3 major areas of application: (1) vascular imaging, for advanced atherosclerotic plaque phenotyping and evaluation of novel imaging tracers or therapeutic interventions; (2) assessment of the ischemic heart and brain; and (3) whole-body imaging of the hematopoietic system. Finally, we will provide insights on potential novel technical developments which may further increase the relevance of integrated positron emission tomography/magnetic resonance imaging in preclinical atherosclerosis studies.
Collapse
Affiliation(s)
- Claudia Calcagno
- From the BioMedical Engineering and Imaging Institute (C.C., C.P.-M., W.J.M.M., Z.A.F.), Icahn School of Medicine at Mount Sinai, NY.,Department of Radiology (C.C., C.P.-M., W.J.M.M., Z.A.F.), Icahn School of Medicine at Mount Sinai, NY
| | - Carlos Pérez-Medina
- From the BioMedical Engineering and Imaging Institute (C.C., C.P.-M., W.J.M.M., Z.A.F.), Icahn School of Medicine at Mount Sinai, NY.,Department of Radiology (C.C., C.P.-M., W.J.M.M., Z.A.F.), Icahn School of Medicine at Mount Sinai, NY.,Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (C.P.-M.)
| | - Willem J M Mulder
- From the BioMedical Engineering and Imaging Institute (C.C., C.P.-M., W.J.M.M., Z.A.F.), Icahn School of Medicine at Mount Sinai, NY.,Department of Radiology (C.C., C.P.-M., W.J.M.M., Z.A.F.), Icahn School of Medicine at Mount Sinai, NY.,Department of Oncological Sciences (W.J.M.M.), Icahn School of Medicine at Mount Sinai, NY.,Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, the Netherlands (W.J.M.M.)
| | - Zahi A Fayad
- From the BioMedical Engineering and Imaging Institute (C.C., C.P.-M., W.J.M.M., Z.A.F.), Icahn School of Medicine at Mount Sinai, NY.,Department of Radiology (C.C., C.P.-M., W.J.M.M., Z.A.F.), Icahn School of Medicine at Mount Sinai, NY
| |
Collapse
|
36
|
Datta P, Ray S. Nanoparticulate formulations of radiopharmaceuticals: Strategy to improve targeting and biodistribution properties. J Labelled Comp Radiopharm 2020; 63:333-355. [PMID: 32220029 DOI: 10.1002/jlcr.3839] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/17/2020] [Accepted: 03/08/2020] [Indexed: 02/06/2023]
Abstract
Application of nanotechnology principles in drug delivery has created opportunities for treatment of several diseases. Nanotechnology offers the advantage of overcoming the adverse biopharmaceutics or pharmacokinetic properties of drug molecules, to be determined by the transport properties of the particles themselves. Through the manipulation of size, shape, charge, and type of nanoparticle delivery system, variety of distribution profiles may be obtained. However, there still exists greater need to derive and standardize definitive structure property relationships for the distribution profiles of the delivery system. When applied to radiopharmaceuticals, the delivery systems assume greater significance. For the safety and efficacy of both diagnostics and therapeutic radiopharmaceuticals, selective localization in target tissue is even more important. At the same time, the synthesis and fabrication reactions of radiolabelled nanoparticles need to be completed in much shorter time. Moreover, the extensive understanding of the several interesting optical and magnetic properties of materials in nanoscale provides for achieving multiple objectives in nuclear medicine. This review discusses the various nanoparticle systems, which are applied for radionuclides and analyses the important bottlenecks that are required to be overcome for their more widespread clinical adaptation.
Collapse
Affiliation(s)
- Pallab Datta
- Centre for Healthcare Science and Technology, Indian Institute of Engineering Science and Technology Shibpur, Howrah, India
| | | |
Collapse
|
37
|
Pérez-Medina C, Fayad ZA, Mulder WJM. Atherosclerosis Immunoimaging by Positron Emission Tomography. Arterioscler Thromb Vasc Biol 2020; 40:865-873. [PMID: 32078338 DOI: 10.1161/atvbaha.119.313455] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The immune system's role in atherosclerosis has long been an important research topic and is increasingly investigated for therapeutic and diagnostic purposes. Therefore, noninvasive imaging of hematopoietic organs and immune cells will undoubtedly improve atherosclerosis phenotyping and serve as a monitoring method for immunotherapeutic treatments. Among the available imaging techniques, positron emission tomography's unique features make it an ideal tool to quantitatively image the immune response in the context of atherosclerosis and afford reliable readouts to guide medical interventions in cardiovascular disease. Here, we summarize the state of the art in the field of atherosclerosis positron emission tomography immunoimaging and provide an outlook on current and future applications.
Collapse
Affiliation(s)
- Carlos Pérez-Medina
- From the Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (C.P.-M.).,Icahn School of Medicine at Mount Sinai, New York (C.P.-M., Z.A.F., W.J.M.M.)
| | - Zahi A Fayad
- Icahn School of Medicine at Mount Sinai, New York (C.P.-M., Z.A.F., W.J.M.M.)
| | - Willem J M Mulder
- Icahn School of Medicine at Mount Sinai, New York (C.P.-M., Z.A.F., W.J.M.M.).,Eindhoven University of Technology, the Netherlands (W.J.M.M.)
| |
Collapse
|
38
|
Mulder WJM, Ochando J, Joosten LAB, Fayad ZA, Netea MG. Therapeutic targeting of trained immunity. Nat Rev Drug Discov 2020; 18:553-566. [PMID: 30967658 DOI: 10.1038/s41573-019-0025-4] [Citation(s) in RCA: 263] [Impact Index Per Article: 65.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Immunotherapy is revolutionizing the treatment of diseases in which dysregulated immune responses have an important role. However, most of the immunotherapy strategies currently being developed engage the adaptive immune system. In the past decade, both myeloid (monocytes, macrophages and dendritic cells) and lymphoid (natural killer cells and innate lymphoid cells) cell populations of the innate immune system have been shown to display long-term changes in their functional programme through metabolic and epigenetic programming. Such reprogramming causes these cells to be either hyperresponsive or hyporesponsive, resulting in a changed immune response to secondary stimuli. This de facto innate immune memory, which has been termed 'trained immunity', provides a powerful 'targeting framework' to regulate the delicate balance of immune homeostasis, priming, training and tolerance. In this Opinion article, we set out our vision of how to target innate immune cells and regulate trained immunity to achieve long-term therapeutic benefits in a range of immune-related diseases. These include conditions characterized by excessive trained immunity, such as inflammatory and autoimmune disorders, allergies and cardiovascular disease and conditions driven by defective trained immunity, such as cancer and certain infections.
Collapse
Affiliation(s)
- Willem J M Mulder
- Translational and Molecular Imaging Institute, Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands. .,Department of Medical Biochemistry, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, Netherlands.
| | - Jordi Ochando
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Transplant Immunology Unit, National Centre of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands.,Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Zahi A Fayad
- Translational and Molecular Imaging Institute, Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands. .,Department for Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany.
| |
Collapse
|
39
|
Chen J, Zhang X, Millican R, Creutzmann JE, Martin S, Jun HW. High density lipoprotein mimicking nanoparticles for atherosclerosis. NANO CONVERGENCE 2020; 7:6. [PMID: 31984429 PMCID: PMC6983461 DOI: 10.1186/s40580-019-0214-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 12/27/2019] [Indexed: 06/10/2023]
Abstract
Atherosclerosis is a major contributor to many cardiovascular events, including myocardial infarction, ischemic stroke, and peripheral arterial disease, making it the leading cause of death worldwide. High-density lipoproteins (HDL), also known as "good cholesterol", have been shown to demonstrate anti-atherosclerotic efficacy through the removal of cholesterol from foam cells in atherosclerotic plaques. Because of the excellent anti-atherosclerotic properties of HDL, in the past several years, there has been tremendous attention in designing HDL mimicking nanoparticles (NPs) of varying functions to image, target, and treat atherosclerosis. In this review, we are summarizing the recent progress in the development of HDL mimicking NPs and their applications for atherosclerosis.
Collapse
Affiliation(s)
- Jun Chen
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL USA
| | - Xixi Zhang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL USA
| | - Reid Millican
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL USA
| | - Jacob Emil Creutzmann
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL USA
| | - Sean Martin
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL USA
| | - Ho-Wook Jun
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL USA
| |
Collapse
|
40
|
Pérez-Medina C, Teunissen AJ, Kluza E, Mulder WJ, van der Meel R. Nuclear imaging approaches facilitating nanomedicine translation. Adv Drug Deliv Rev 2020; 154-155:123-141. [PMID: 32721459 DOI: 10.1016/j.addr.2020.07.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/08/2020] [Accepted: 07/17/2020] [Indexed: 02/07/2023]
Abstract
Nanomedicine approaches can effectively modulate the biodistribution and bioavailability of therapeutic agents, improving their therapeutic index. However, despite the ever-increasing amount of literature reporting on preclinical nanomedicine, the number of nanotherapeutics receiving FDA approval remains relatively low. Several barriers exist that hamper the effective preclinical evaluation and clinical translation of nanotherapeutics. Key barriers include insufficient understanding of nanomedicines' in vivo behavior, inadequate translation from murine models to larger animals, and a lack of patient stratification strategies. Integrating quantitative non-invasive imaging techniques in nanomedicine development offers attractive possibilities to address these issues. Among the available imaging techniques, nuclear imaging by positron emission tomography (PET) and single-photon emission computed tomography (SPECT) are highly attractive in this context owing to their quantitative nature and uncontested sensitivity. In basic and translational research, nuclear imaging techniques can provide critical quantitative information about pharmacokinetic parameters, biodistribution profiles or target site accumulation of nanocarriers and their associated payload. During clinical evaluation, nuclear imaging can be used to select patients amenable to nanomedicine treatment. Here, we review how nuclear imaging-based approaches are increasingly being integrated into nanomedicine development and discuss future developments that will accelerate their clinical translation.
Collapse
|
41
|
Beldman T, Malinova TS, Desclos E, Grootemaat AE, Misiak ALS, van der Velden S, van Roomen CPAA, Beckers L, van Veen HA, Krawczyk PM, Hoebe RA, Sluimer JC, Neele AE, de Winther MPJ, van der Wel NN, Lutgens E, Mulder WJM, Huveneers S, Kluza E. Nanoparticle-Aided Characterization of Arterial Endothelial Architecture during Atherosclerosis Progression and Metabolic Therapy. ACS NANO 2019; 13:13759-13774. [PMID: 31268670 PMCID: PMC6933811 DOI: 10.1021/acsnano.8b08875] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 07/03/2019] [Indexed: 05/08/2023]
Abstract
Atherosclerosis is associated with a compromised endothelial barrier, facilitating the accumulation of immune cells and macromolecules in atherosclerotic lesions. In this study, we investigate endothelial barrier integrity and the enhanced permeability and retention (EPR) effect during atherosclerosis progression and therapy in Apoe-/- mice using hyaluronan nanoparticles (HA-NPs). Utilizing ultrastructural and en face plaque imaging, we uncover a significantly decreased junction continuity in the atherosclerotic plaque-covering endothelium compared to the normal vessel wall, indicative of disrupted endothelial barrier. Intriguingly, the plaque advancement had a positive effect on junction stabilization, which correlated with a 3-fold lower accumulation of in vivo administrated HA-NPs in advanced plaques compared to early counterparts. Furthermore, by using super-resolution and correlative light and electron microscopy, we trace nanoparticles in the plaque microenvironment. We find nanoparticle-enriched endothelial junctions, containing 75% of detected HA-NPs, and a high HA-NP accumulation in the endothelium-underlying extracellular matrix, which suggest an endothelial junctional traffic of HA-NPs to the plague. Finally, we probe the EPR effect by HA-NPs in the context of metabolic therapy with a glycolysis inhibitor, 3PO, proposed as a vascular normalizing strategy. The observed trend of attenuated HA-NP uptake in aortas of 3PO-treated mice coincides with the endothelial silencing activity of 3PO, demonstrated in vitro. Interestingly, the therapy also reduced the plaque inflammatory burden, while activating macrophage metabolism. Our findings shed light on natural limitations of nanoparticle accumulation in atherosclerotic plaques and provide mechanistic insight into nanoparticle trafficking across the atherosclerotic endothelium. Furthermore, our data contribute to the rising field of endothelial barrier modulation in atherosclerosis.
Collapse
Affiliation(s)
- Thijs
J. Beldman
- Experimental
Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular
Sciences (ACS), Amsterdam University Medical
Center, Amsterdam 1105 AZ, The Netherlands
| | - Tsveta S. Malinova
- Vascular
Microenvironment and Integrity, Department of Medical Biochemistry,
Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Center, Amsterdam 1105 AZ, The
Netherlands
| | - Emilie Desclos
- Cellular
Imaging-Core Facility, Academic Medical
Center, Amsterdam 1105 AZ, The Netherlands
| | - Anita E. Grootemaat
- Cellular
Imaging-Core Facility, Academic Medical
Center, Amsterdam 1105 AZ, The Netherlands
| | - Aresh L. S. Misiak
- Experimental
Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular
Sciences (ACS), Amsterdam University Medical
Center, Amsterdam 1105 AZ, The Netherlands
| | - Saskia van der Velden
- Experimental
Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular
Sciences (ACS), Amsterdam University Medical
Center, Amsterdam 1105 AZ, The Netherlands
| | - Cindy P. A. A. van Roomen
- Experimental
Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular
Sciences (ACS), Amsterdam University Medical
Center, Amsterdam 1105 AZ, The Netherlands
| | - Linda Beckers
- Experimental
Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular
Sciences (ACS), Amsterdam University Medical
Center, Amsterdam 1105 AZ, The Netherlands
| | - Henk A. van Veen
- Cellular
Imaging-Core Facility, Academic Medical
Center, Amsterdam 1105 AZ, The Netherlands
| | - Przemyslaw M. Krawczyk
- Department
of Medical Biology, Amsterdam University
Medical Center, Amsterdam 1105 AZ, The Netherlands
| | - Ron A. Hoebe
- Cellular
Imaging-Core Facility, Academic Medical
Center, Amsterdam 1105 AZ, The Netherlands
| | - Judith C. Sluimer
- Department
of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht 6229 ER, The Netherlands
| | - Annette E. Neele
- Experimental
Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular
Sciences (ACS), Amsterdam University Medical
Center, Amsterdam 1105 AZ, The Netherlands
| | - Menno P. J. de Winther
- Experimental
Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular
Sciences (ACS), Amsterdam University Medical
Center, Amsterdam 1105 AZ, The Netherlands
- Institute
for Cardiovascular Prevention, Ludwig Maximilians
University, Munich 80336, Germany
| | - Nicole N. van der Wel
- Cellular
Imaging-Core Facility, Academic Medical
Center, Amsterdam 1105 AZ, The Netherlands
| | - Esther Lutgens
- Experimental
Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular
Sciences (ACS), Amsterdam University Medical
Center, Amsterdam 1105 AZ, The Netherlands
- Institute
for Cardiovascular Prevention, Ludwig Maximilians
University, Munich 80336, Germany
| | - Willem J. M. Mulder
- Experimental
Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular
Sciences (ACS), Amsterdam University Medical
Center, Amsterdam 1105 AZ, The Netherlands
- Translational
and Molecular Imaging Institute, Icahn School
of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Stephan Huveneers
- Vascular
Microenvironment and Integrity, Department of Medical Biochemistry,
Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Center, Amsterdam 1105 AZ, The
Netherlands
| | - Ewelina Kluza
- Experimental
Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular
Sciences (ACS), Amsterdam University Medical
Center, Amsterdam 1105 AZ, The Netherlands
| |
Collapse
|
42
|
Cicha I, Chauvierre C, Texier I, Cabella C, Metselaar JM, Szebeni J, Dézsi L, Alexiou C, Rouzet F, Storm G, Stroes E, Bruce D, MacRitchie N, Maffia P, Letourneur D. From design to the clinic: practical guidelines for translating cardiovascular nanomedicine. Cardiovasc Res 2019; 114:1714-1727. [PMID: 30165574 DOI: 10.1093/cvr/cvy219] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 08/23/2018] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular diseases (CVD) account for nearly half of all deaths in Europe and almost 30% of global deaths. Despite the improved clinical management, cardiovascular mortality is predicted to rise in the next decades due to the increasing impact of aging, obesity, and diabetes. The goal of emerging cardiovascular nanomedicine is to reduce the burden of CVD using nanoscale medical products and devices. However, the development of novel multicomponent nano-sized products poses multiple technical, ethical, and regulatory challenges, which often obstruct their road to successful approval and use in clinical practice. This review discusses the rational design of nanoparticles, including safety considerations and regulatory issues, and highlights the steps needed to achieve efficient clinical translation of promising nanomedicinal products for cardiovascular applications.
Collapse
Affiliation(s)
- Iwona Cicha
- Cardiovascular Nanomedicine Unit, Section of Experimental Oncology und Nanomedicine (SEON), ENT-Department, University Hospital Erlangen, Glückstr. 10a, Erlangen, Germany
| | - Cédric Chauvierre
- INSERM U1148, LVTS, Paris Diderot University, Paris 13 University, X. Bichat Hospital, 46 rue H. Huchard, Paris, France
| | | | - Claudia Cabella
- Centro Ricerche Bracco, Bracco Imaging Spa, Colleretto Giacosa, Italy
| | - Josbert M Metselaar
- Department of Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH-Aachen University, Aachen, Germany
| | - János Szebeni
- Nanomedicine Research and Education Center, Department of Pathophysiology, Semmelweis University, Budapest, Hungary
| | - László Dézsi
- Nanomedicine Research and Education Center, Department of Pathophysiology, Semmelweis University, Budapest, Hungary
| | - Christoph Alexiou
- Cardiovascular Nanomedicine Unit, Section of Experimental Oncology und Nanomedicine (SEON), ENT-Department, University Hospital Erlangen, Glückstr. 10a, Erlangen, Germany
| | - François Rouzet
- INSERM U1148, LVTS, Paris Diderot University, Paris 13 University, X. Bichat Hospital, 46 rue H. Huchard, Paris, France.,Department of Nuclear Medicine, X. Bichat Hospital, Paris, France
| | - Gert Storm
- Department of Pharmaceutics, University of Utrecht, Utrecht, The Netherlands.,Department of Biomaterials Science and Technology, University of Twente, Enschede, The Netherlands
| | - Erik Stroes
- Department of Vascular Medicine, Amsterdam Medical Center, Amsterdam, The Netherlands
| | | | - Neil MacRitchie
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Pasquale Maffia
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.,Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.,Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Didier Letourneur
- INSERM U1148, LVTS, Paris Diderot University, Paris 13 University, X. Bichat Hospital, 46 rue H. Huchard, Paris, France
| |
Collapse
|
43
|
Rodell CB, Koch PD, Weissleder R. Screening for new macrophage therapeutics. Theranostics 2019; 9:7714-7729. [PMID: 31695796 PMCID: PMC6831478 DOI: 10.7150/thno.34421] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/19/2019] [Indexed: 12/15/2022] Open
Abstract
Myeloid derived macrophages play a key role in many human diseases, and their therapeutic modulation via pharmacological means is receiving considerable attention. Of particular interest is the fact that these cells are i) dynamic phenotypes well suited to therapeutic manipulation and ii) phagocytic, allowing them to be efficiently targeted with nanoformulations. However, it is important to consider that macrophages represent heterogeneous populations of subtypes with often competing biological behaviors and functions. In order to develop next generation therapeutics, it is therefore essential to screen for biological effects through a combination of in vitro and in vivo assays. Here, we review the state-of-the-art techniques, including both cell based screens and in vivo imaging tools that have been developed for assessment of macrophage phenotype. We conclude with a forward-looking perspective on the growing need for noninvasive macrophage assessment and laboratory assays to be put into clinical practice and the potential broader impact of myeloid-targeted therapeutics.
Collapse
|
44
|
Binderup T, Duivenvoorden R, Fay F, van Leent MMT, Malkus J, Baxter S, Ishino S, Zhao Y, Sanchez-Gaytan B, Teunissen AJP, Frederico YCA, Tang J, Carlucci G, Lyashchenko S, Calcagno C, Karakatsanis N, Soultanidis G, Senders ML, Robson PM, Mani V, Ramachandran S, Lobatto ME, Hutten BA, Granada JF, Reiner T, Swirski FK, Nahrendorf M, Kjaer A, Fisher EA, Fayad ZA, Pérez-Medina C, Mulder WJM. Imaging-assisted nanoimmunotherapy for atherosclerosis in multiple species. Sci Transl Med 2019; 11:eaaw7736. [PMID: 31434756 PMCID: PMC7328283 DOI: 10.1126/scitranslmed.aaw7736] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 07/09/2019] [Indexed: 01/01/2023]
Abstract
Nanomedicine research produces hundreds of studies every year, yet very few formulations have been approved for clinical use. This is due in part to a reliance on murine studies, which have limited value in accurately predicting translational efficacy in larger animal models and humans. Here, we report the scale-up of a nanoimmunotherapy from mouse to large rabbit and porcine atherosclerosis models, with an emphasis on the solutions we implemented to overcome production and evaluation challenges. Specifically, we integrated translational imaging readouts within our workflow to both analyze the nanoimmunotherapeutic's in vivo behavior and assess treatment response in larger animals. We observed our nanoimmunotherapeutic's anti-inflammatory efficacy in mice, as well as rabbits and pigs. Nanoimmunotherapy-mediated reduction of inflammation in the large animal models halted plaque progression, supporting the approach's translatability and potential to acutely treat atherosclerosis.
Collapse
Affiliation(s)
- Tina Binderup
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Clinical Physiology, Nuclear Medicine and PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, 2100 Copenhagen, Denmark
| | - Raphaël Duivenvoorden
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 XZ Nijmegen, Netherlands
| | - Francois Fay
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Institut Galien Paris Sud, Faculté de Pharmacie, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France
| | - Mandy M T van Leent
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Joost Malkus
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Samantha Baxter
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Seigo Ishino
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yiming Zhao
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Brenda Sanchez-Gaytan
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Abraham J P Teunissen
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yohana C A Frederico
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jun Tang
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Giuseppe Carlucci
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Bernard and Irene Schwarz Center for Biomedical Imaging, New York University, New York, NY 10016, USA
| | - Serge Lyashchenko
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Radiology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Claudia Calcagno
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nicolas Karakatsanis
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Georgios Soultanidis
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Max L Senders
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Medical Biochemistry, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, Netherlands
| | - Philip M Robson
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Venkatesh Mani
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sarayu Ramachandran
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mark E Lobatto
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Radiology, Spaarne Gasthuis, 2035 RC Haarlem, Netherlands
| | - Barbara A Hutten
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam University Medical Centers, 1105 AZ Amsterdam, Netherlands
| | - Juan F Granada
- CRF Skirball Center for Innovation, Cardiovascular Research Foundation, Orangeburg, NY 10962, USA
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Radiology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Filip K Swirski
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Matthias Nahrendorf
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine and PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, 2100 Copenhagen, Denmark
| | - Edward A Fisher
- Department of Medicine (Cardiology) and Cell Biology, Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Zahi A Fayad
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Carlos Pérez-Medina
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain
| | - Willem J M Mulder
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
- Department of Medical Biochemistry, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, Netherlands
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, 5612 AZ Eindhoven, Netherlands
| |
Collapse
|
45
|
Mason CA, Kossatz S, Carter LM, Pirovano G, Brand C, Guru N, Pérez-Medina C, Lewis JS, Mulder WJM, Reiner T. An 89Zr-HDL PET Tracer Monitors Response to a CSF1R Inhibitor. J Nucl Med 2019; 61:433-436. [PMID: 31420495 PMCID: PMC7067531 DOI: 10.2967/jnumed.119.230466] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 07/24/2019] [Indexed: 12/11/2022] Open
Abstract
The immune function within the tumor microenvironment has become a prominent therapeutic target, with tumor-associated macrophages (TAMs) playing a critical role in immune suppression. We propose an 89Zr-labeled high-density lipoprotein (89Zr-HDL) nanotracer as a means of monitoring response to immunotherapy. Methods: Female MMTV-PyMT mice were treated with pexidartinib, a colony-stimulating factor 1 receptor (CSF1R) inhibitor, to reduce TAM density. The accumulation of 89Zr-HDL within the tumor was assessed using PET/CT imaging and autoradiography, whereas TAM burden was determined using immunofluorescence. Results: A significant reduction in 89Zr-HDL accumulation was observed in PET/CT images, with 2.9% ± 0.3% and 3.7% ± 0.2% injected dose/g for the pexidartinib- and vehicle-treated mice, respectively. This reduction was corroborated ex vivo and correlated with decreased TAM density. Conclusion: These results support the potential use of 89Zr-HDL nanoparticles as a PET tracer to quickly monitor the response to CSF1R inhibitors and other therapeutic strategies targeting TAMs.
Collapse
Affiliation(s)
- Christian A Mason
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Susanne Kossatz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lukas M Carter
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Giacomo Pirovano
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Christian Brand
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Navjot Guru
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Carlos Pérez-Medina
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Willem J M Mulder
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.,Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands.,Department of Medical Biochemistry, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, The Netherlands
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York .,Department of Radiology, Weill Cornell Medical College, New York, New York.,Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York; and.,Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
46
|
Yong-Sang J, Dioury F, Meneyrol V, Ait-Arsa I, Idoumbin JP, Guibbal F, Patché J, Gimié F, Khantalin I, Couprie J, Giraud P, Benard S, Ferroud C, Jestin E, Meilhac O. Development, synthesis, and 68Ga-Labeling of a Lipophilic complexing agent for atherosclerosis PET imaging. Eur J Med Chem 2019; 176:129-134. [PMID: 31102933 DOI: 10.1016/j.ejmech.2019.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/05/2019] [Accepted: 05/01/2019] [Indexed: 10/26/2022]
Abstract
Cardiovascular disease is the leading cause of mortality and morbidity worldwide. Atherosclerosis accounts for 50% of deaths in western countries. This multifactorial pathology is characterized by the accumulation of lipids and inflammatory cells within the vascular wall, leading to plaque formation. We describe herein the synthesis of a PCTA-based 68Ga3+ chelator coupled to a phospholipid biovector 1,2-distearoyl-sn-glycero-3-phosphoethanolamine (DSPE), which is the main constituent of the phospholipid moiety of High-Density Lipoprotein (HDL) phospholipid moiety. The resulting 68Ga-PCTA-DSPE inserted into HDL particles was compared to 18F-FDG as a PET agent to visualize atherosclerotic plaques. Our agent markedly accumulated within mouse atheromatous aortas and more interestingly in human endarterectomy carotid samples. These results support the potential use of 68Ga-PCTA-DSPE-HDL for atherosclerosis PET imaging.
Collapse
Affiliation(s)
- Jennyfer Yong-Sang
- Université de La Réunion, Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Plateforme CYROI, 2 rue Maxime Rivière, 97490, Sainte-Clotilde, Réunion, France; Laboratoire de Génomique, Bioinformatique, et Chimie Moléculaire, EA 7528, Conservatoire national des Arts et métiers, 2 rue Conté, 75003, Paris, HESAM Université, France
| | - Fabienne Dioury
- Laboratoire de Génomique, Bioinformatique, et Chimie Moléculaire, EA 7528, Conservatoire national des Arts et métiers, 2 rue Conté, 75003, Paris, HESAM Université, France
| | - Vincent Meneyrol
- Groupe d'Intérêt Public, Cyclotron Réunion Océan Indien, 2 rue Maxime Rivière, 97490, Sainte-Clotilde, Réunion, France
| | - Imade Ait-Arsa
- Groupe d'Intérêt Public, Cyclotron Réunion Océan Indien, 2 rue Maxime Rivière, 97490, Sainte-Clotilde, Réunion, France
| | - Jean-Patrick Idoumbin
- Groupe d'Intérêt Public, Cyclotron Réunion Océan Indien, 2 rue Maxime Rivière, 97490, Sainte-Clotilde, Réunion, France
| | - Florian Guibbal
- Université de La Réunion, Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Plateforme CYROI, 2 rue Maxime Rivière, 97490, Sainte-Clotilde, Réunion, France
| | - Jessica Patché
- Université de La Réunion, Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Plateforme CYROI, 2 rue Maxime Rivière, 97490, Sainte-Clotilde, Réunion, France
| | - Fanny Gimié
- Groupe d'Intérêt Public, Cyclotron Réunion Océan Indien, 2 rue Maxime Rivière, 97490, Sainte-Clotilde, Réunion, France
| | - Ilya Khantalin
- Université de La Réunion, Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Plateforme CYROI, 2 rue Maxime Rivière, 97490, Sainte-Clotilde, Réunion, France; CHU de La Réunion, Allée des Topazes, 97400, Saint-Denis, Réunion, France
| | - Joël Couprie
- Université de La Réunion, Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Plateforme CYROI, 2 rue Maxime Rivière, 97490, Sainte-Clotilde, Réunion, France
| | - Pierre Giraud
- Université de La Réunion, Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Plateforme CYROI, 2 rue Maxime Rivière, 97490, Sainte-Clotilde, Réunion, France
| | - Sébastien Benard
- Groupe d'Intérêt Public, Cyclotron Réunion Océan Indien, 2 rue Maxime Rivière, 97490, Sainte-Clotilde, Réunion, France
| | - Clotilde Ferroud
- Laboratoire de Génomique, Bioinformatique, et Chimie Moléculaire, EA 7528, Conservatoire national des Arts et métiers, 2 rue Conté, 75003, Paris, HESAM Université, France
| | - Emmanuelle Jestin
- Université de La Réunion, Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Plateforme CYROI, 2 rue Maxime Rivière, 97490, Sainte-Clotilde, Réunion, France; Groupe d'Intérêt Public, Cyclotron Réunion Océan Indien, 2 rue Maxime Rivière, 97490, Sainte-Clotilde, Réunion, France
| | - Olivier Meilhac
- Université de La Réunion, Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Plateforme CYROI, 2 rue Maxime Rivière, 97490, Sainte-Clotilde, Réunion, France; CHU de La Réunion, Allée des Topazes, 97400, Saint-Denis, Réunion, France.
| |
Collapse
|
47
|
PET/MR Imaging of Malondialdehyde-Acetaldehyde Epitopes With a Human Antibody Detects Clinically Relevant Atherothrombosis. J Am Coll Cardiol 2019; 71:321-335. [PMID: 29348025 DOI: 10.1016/j.jacc.2017.11.036] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 10/22/2017] [Accepted: 11/06/2017] [Indexed: 12/30/2022]
Abstract
BACKGROUND Oxidation-specific epitopes (OSEs) are proinflammatory, and elevated levels in plasma predict cardiovascular events. OBJECTIVES The purpose of this study was to develop novel positron emission tomography (PET) probes to noninvasively image OSE-rich lesions. METHODS An antigen-binding fragment (Fab) antibody library was constructed from human fetal cord blood. After multiple rounds of screening against malondialdehyde-acetaldehyde (MAA) epitopes, the Fab LA25 containing minimal nontemplated insertions in the CDR3 region was identified and characterized. In mice, pharmacokinetics, biodistribution, and plaque specificity studies were performed with Zirconium-89 (89Zr)-labeled LA25. In rabbits, 89Zr-LA25 was used in combination with an integrated clinical PET/magnetic resonance (MR) system. 18F-fluorodeoxyglucose PET and dynamic contrast-enhanced MR imaging were used to evaluate vessel wall inflammation and plaque neovascularization, respectively. Extensive ex vivo validation was carried out through a combination of gamma counting, near infrared fluorescence, autoradiography, immunohistochemistry, and immunofluorescence. RESULTS LA25 bound specifically to MAA epitopes in advanced and ruptured human atherosclerotic plaques with accompanying thrombi and in debris from distal protection devices. PET/MR imaging 24 h after injection of 89Zr-LA25 showed increased uptake in the abdominal aorta of atherosclerotic rabbits compared with nonatherosclerotic control rabbits, confirmed by ex vivo gamma counting and autoradiography. 18F-fluorodeoxyglucose PET, dynamic contrast-enhanced MR imaging, and near-infrared fluorescence signals were also significantly higher in atherosclerotic rabbit aortas compared with control aortas. Enhanced liver uptake was also noted in atherosclerotic animals, confirmed by the presence of MAA epitopes by immunostaining. CONCLUSIONS 89Zr-LA25 is a novel PET radiotracer that may allow noninvasive phenotyping of high-risk OSE-rich lesions.
Collapse
|
48
|
Lobatto ME, Binderup T, Robson PM, Giesen LFP, Calcagno C, Witjes J, Fay F, Baxter S, Wessel CH, Eldib M, Bini J, Carlin SD, Stroes ESG, Storm G, Kjaer A, Lewis JS, Reiner T, Fayad ZA, Mulder WJM, Pérez-Medina C. Multimodal Positron Emission Tomography Imaging to Quantify Uptake of 89Zr-Labeled Liposomes in the Atherosclerotic Vessel Wall. Bioconjug Chem 2019; 31:360-368. [PMID: 31095372 DOI: 10.1021/acs.bioconjchem.9b00256] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nanotherapy has recently emerged as an experimental treatment option for atherosclerosis. To fulfill its promise, robust noninvasive imaging approaches for subject selection and treatment evaluation are warranted. To that end, we present here a positron emission tomography (PET)-based method for quantification of liposomal nanoparticle uptake in the atherosclerotic vessel wall. We evaluated a modular procedure to label liposomal nanoparticles with the radioisotope zirconium-89 (89Zr). Their biodistribution and vessel wall targeting in a rabbit atherosclerosis model was evaluated up to 15 days after intravenous injection by PET/computed tomography (CT) and PET/magnetic resonance imaging (PET/MRI). Vascular permeability was assessed in vivo using three-dimensional dynamic contrast-enhanced MRI (3D DCE-MRI) and ex vivo using near-infrared fluorescence (NIRF) imaging. The 89Zr-radiolabeled liposomes displayed a biodistribution pattern typical of long-circulating nanoparticles. Importantly, they markedly accumulated in atherosclerotic lesions in the abdominal aorta, as evident on PET/MRI and confirmed by autoradiography, and this uptake moderately correlated with vascular permeability. The method presented herein facilitates the development of nanotherapy for atherosclerotic disease as it provides a tool to screen for nanoparticle targeting in individual subjects' plaques.
Collapse
Affiliation(s)
- Mark E Lobatto
- Translational and Molecular Imaging Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States.,Department of Radiology , Spaarne Gasthuis , 2035 RC Haarlem , The Netherlands
| | - Tina Binderup
- Translational and Molecular Imaging Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States.,Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging , Rigshospitalet & University of Copenhagen , 2100 Copenhagen , Denmark
| | - Philip M Robson
- Translational and Molecular Imaging Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Luuk F P Giesen
- Translational and Molecular Imaging Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Claudia Calcagno
- Translational and Molecular Imaging Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Julia Witjes
- Translational and Molecular Imaging Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Francois Fay
- Translational and Molecular Imaging Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States.,Institut Galien Paris Sud UMR 8612, Faculté de Pharmacie, CNRS, Univ. Paris-Sud Université Paris-Saclay , 92290 Châtenay-Malabry , France
| | - Samantha Baxter
- Translational and Molecular Imaging Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Chang Ho Wessel
- Translational and Molecular Imaging Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Mootaz Eldib
- Translational and Molecular Imaging Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Jason Bini
- Translational and Molecular Imaging Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Sean D Carlin
- Department of Radiology , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States
| | - Erik S G Stroes
- Department of Vascular Medicine , Academic Medical Center , 1105 AZ Amsterdam , The Netherlands
| | - Gert Storm
- Department of Targeted Therapeutics, MIRA Institute , University of Twente , 7522 NB Enschede , The Netherlands.,Utrecht Institute for Pharmaceutical Sciences , Utrecht University , 3512 JE Utrecht , The Netherlands
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging , Rigshospitalet & University of Copenhagen , 2100 Copenhagen , Denmark
| | - Jason S Lewis
- Department of Radiology , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States.,Program in Molecular Pharmacology , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States
| | - Thomas Reiner
- Department of Radiology , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States.,Chemical Biology Program , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States
| | - Zahi A Fayad
- Translational and Molecular Imaging Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Willem J M Mulder
- Translational and Molecular Imaging Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States.,Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems , Eindhoven University of Technology , 5612 AZ Eindhoven , The Netherlands.,Department of Oncological Sciences , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Carlos Pérez-Medina
- Translational and Molecular Imaging Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States.,Centro Nacional de Investigaciones Cardiovasculares Carlos III , 28029 Madrid , Spain
| |
Collapse
|
49
|
Woodside DG. Nanoparticle Imaging of Vascular Inflammation and Remodeling in Atherosclerotic Disease. CURRENT CARDIOVASCULAR IMAGING REPORTS 2019. [DOI: 10.1007/s12410-019-9501-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
50
|
Ahmed M, Baumgartner R, Aldi S, Dusart P, Hedin U, Gustafsson B, Caidahl K. Human serum albumin-based probes for molecular targeting of macrophage scavenger receptors. Int J Nanomedicine 2019; 14:3723-3741. [PMID: 31190821 PMCID: PMC6535103 DOI: 10.2147/ijn.s197990] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 03/01/2019] [Indexed: 02/05/2023] Open
Abstract
Background: Inflammation and accumulation of macrophages are key features of unstable atherosclerotic plaques. The ability of macrophages to take up molecular probes can be exploited in new clinical imaging methods for the detection of unstable atherosclerotic lesions. We investigated whether modifications of human serum albumin (HSA) could be used to target macrophages efficiently in vitro. Materials and methods: Maleylated and aconitylated HSA were compared with unmodified HSA. Fluorescent or radiolabeled (89Zr) modified HSA was used in in vitro experiments to study cellular uptake by differentiated THP-1 cells and primary human macrophages. The time course of uptake was evaluated by flow cytometry, confocal microscopy, real-time microscopy and radioactivity measurements. The involvement of scavenger receptors (SR-A1, SR-B2, LOX-1) was assessed by knockdown experiments using RNA interference, by blocking experiments and by assays of competition by modified low-density lipoprotein. Results: Modified HSA was readily taken up by different macrophages. Uptake was mediated nonexclusively via the scavenger receptor SR-A1 (encoded by the MSR1 gene). Knockdown of CD36 and ORL1 had no influence on the uptake. Modified HSA was preferentially taken up by human macrophages compared with other vascular cell types such as endothelial cells and smooth muscle cells. Conclusions: Modified 89Zr-labeled HSA probes were recognized by different subsets of polarized macrophages, and maleylated HSA may be a promising radiotracer for radionuclide imaging of macrophage-rich inflammatory vascular diseases.
Collapse
Affiliation(s)
- Mona Ahmed
- Department of Molecular Medicine and Surgery, Karolinska Institutet, SE 17176, Stockholm, Sweden
| | - Roland Baumgartner
- Department of Medicine Solna, Karolinska Institutet, SE 17176, Stockholm, Sweden
| | - Silvia Aldi
- Section for Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE 17177, Stockholm, Sweden
| | - Philip Dusart
- Department of Cellular and Clinical Proteomics, Science for Life Laboratory, Kungliga Tekniska Högskolan (KTH), SE 17165, Stockholm, Sweden
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska Institutet, SE 17176, Stockholm, Sweden
| | - Björn Gustafsson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, SE 17176, Stockholm, Sweden
| | - Kenneth Caidahl
- Department of Molecular Medicine and Surgery, Karolinska Institutet, SE 17176, Stockholm, Sweden
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE 41345, Gothenburg, Sweden
| |
Collapse
|