101
|
Protein MRI contrast agent with unprecedented metal selectivity and sensitivity for liver cancer imaging. Proc Natl Acad Sci U S A 2015; 112:6607-12. [PMID: 25971726 DOI: 10.1073/pnas.1423021112] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
With available MRI techniques, primary and metastatic liver cancers that are associated with high mortality rates and poor treatment responses are only diagnosed at late stages, due to the lack of highly sensitive contrast agents without Gd(3+) toxicity. We have developed a protein contrast agent (ProCA32) that exhibits high stability for Gd(3+) and a 10(11)-fold greater selectivity for Gd(3+) over Zn(2+) compared with existing contrast agents. ProCA32, modified from parvalbumin, possesses high relaxivities (r1/r2: 66.8 mmol(-1)⋅s(-1)/89.2 mmol(-1)⋅s(-1) per particle). Using T1- and T2-weighted, as well as T2/T1 ratio imaging, we have achieved, for the first time (to our knowledge), robust MRI detection of early liver metastases as small as ∼0.24 mm in diameter, much smaller than the current detection limit of 10-20 mm. Furthermore, ProCA32 exhibits appropriate in vivo preference for liver sinusoidal spaces and pharmacokinetics for high-quality imaging. ProCA32 will be invaluable for noninvasive early detection of primary and metastatic liver cancers as well as for monitoring treatment and guiding therapeutic interventions, including drug delivery.
Collapse
|
102
|
|
103
|
Palekar RU, Jallouk AP, Goette MJ, Chen J, Myerson JW, Allen JS, Akk A, Yang L, Tu Y, Miller MJ, Pham CTN, Wickline SA, Pan H. Quantifying progression and regression of thrombotic risk in experimental atherosclerosis. FASEB J 2015; 29:3100-9. [PMID: 25857553 DOI: 10.1096/fj.14-269084] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 03/16/2015] [Indexed: 12/17/2022]
Abstract
Currently, there are no generally applicable noninvasive methods for defining the relationship between atherosclerotic vascular damage and risk of focal thrombosis. Herein, we demonstrate methods to delineate the progression and regression of vascular damage in response to an atherogenic diet by quantifying the in vivo accumulation of semipermeable 200-300 nm perfluorocarbon core nanoparticles (PFC-NP) in ApoE null mouse plaques with [(19)F] magnetic resonance spectroscopy (MRS). Permeability to PFC-NP remained minimal until 12 weeks on diet, then increased rapidly following 12 weeks, but regressed to baseline within 8 weeks after diet normalization. Markedly accelerated clotting (53.3% decrease in clotting time) was observed in carotid artery preparations of fat-fed mice subjected to photochemical injury as defined by the time to flow cessation. For all mice on and off diet, an inverse linear relationship was observed between the permeability to PFC-NP and accelerated thrombosis (P = 0.02). Translational feasibility for quantifying plaque permeability and vascular damage in vivo was demonstrated with clinical 3 T MRI of PFC-NP accumulating in plaques of atherosclerotic rabbits. These observations suggest that excessive permeability to PFC-NP may indicate prothrombotic risk in damaged atherosclerotic vasculature, which resolves within weeks after dietary therapy.
Collapse
Affiliation(s)
- Rohun U Palekar
- *Department of Biomedical Engineering and Department of Medicine, Washington University, St. Louis, Missouri, USA
| | - Andrew P Jallouk
- *Department of Biomedical Engineering and Department of Medicine, Washington University, St. Louis, Missouri, USA
| | - Matthew J Goette
- *Department of Biomedical Engineering and Department of Medicine, Washington University, St. Louis, Missouri, USA
| | - Junjie Chen
- *Department of Biomedical Engineering and Department of Medicine, Washington University, St. Louis, Missouri, USA
| | - Jacob W Myerson
- *Department of Biomedical Engineering and Department of Medicine, Washington University, St. Louis, Missouri, USA
| | - John S Allen
- *Department of Biomedical Engineering and Department of Medicine, Washington University, St. Louis, Missouri, USA
| | - Antonina Akk
- *Department of Biomedical Engineering and Department of Medicine, Washington University, St. Louis, Missouri, USA
| | - Lihua Yang
- *Department of Biomedical Engineering and Department of Medicine, Washington University, St. Louis, Missouri, USA
| | - Yizheng Tu
- *Department of Biomedical Engineering and Department of Medicine, Washington University, St. Louis, Missouri, USA
| | - Mark J Miller
- *Department of Biomedical Engineering and Department of Medicine, Washington University, St. Louis, Missouri, USA
| | - Christine T N Pham
- *Department of Biomedical Engineering and Department of Medicine, Washington University, St. Louis, Missouri, USA
| | - Samuel A Wickline
- *Department of Biomedical Engineering and Department of Medicine, Washington University, St. Louis, Missouri, USA
| | - Hua Pan
- *Department of Biomedical Engineering and Department of Medicine, Washington University, St. Louis, Missouri, USA
| |
Collapse
|
104
|
Magnoni M, Ammirati E, Camici PG. Non-invasive molecular imaging of vulnerable atherosclerotic plaques. J Cardiol 2015; 65:261-9. [PMID: 25702846 DOI: 10.1016/j.jjcc.2015.01.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 12/26/2014] [Indexed: 10/24/2022]
Abstract
The growing discoveries coming from clinical and basic research during the past decades have revolutionized our knowledge regarding pathophysiologic mechanisms underlying the atherosclerotic process and its thrombotic complications. The traditional view focusing on the severity of stenosis of atherosclerotic plaque has given way to the evidence that the clinical complications of atherosclerotic vascular disease, particularly the propensity to develop thrombotic complications, are determined mainly by the biological composition of the plaque. This paradigm shift has reinforced the need to move from the sole anatomical assessment toward combined anatomic and functional imaging modalities enabling the molecular and cellular characterization of the disease on top of its structural properties. Together, the progress to identify molecular targets related to plaque vulnerability and the improvement of imaging techniques for the detection of such molecular targets have allowed us to obtain new important pathophysiological information. This might allow better patient stratification for the identification of subjects at high risk to develop premature atherosclerosis who might need an aggressive therapeutic approach. Nuclear techniques, magnetic resonance imaging, computed tomography angiography, and contrast-enhanced ultrasound represent the currently available non-invasive imaging modalities for molecular imaging which can provide different and complementary insights into the biological features of the atherosclerotic process. This clinical review will discuss the evidence and potential translational applications of the individual imaging techniques particularly concerning their ability to detect the main atherosclerotic features related to plaque vulnerability, such as plaque inflammation and intertwined neovascularization.
Collapse
Affiliation(s)
- Marco Magnoni
- Università Vita-Salute San Raffaele and Department of Cardiothoracic and Vascular Diseases, San Raffaele Scientific Institute, Milan, Italy; Heart Care Foundation, Florence, Italy.
| | - Enrico Ammirati
- Università Vita-Salute San Raffaele and Department of Cardiothoracic and Vascular Diseases, San Raffaele Scientific Institute, Milan, Italy; Cardiovascular and Thoracic Department, AO Niguarda Ca' Granda, Milan, Italy
| | - Paolo G Camici
- Università Vita-Salute San Raffaele and Department of Cardiothoracic and Vascular Diseases, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
105
|
Wang K, Pan D, Schmieder AH, Senpan A, Caruthers SD, Cui G, Allen JS, Zhang H, Shen B, Lanza GM. Atherosclerotic neovasculature MR imaging with mixed manganese-gadolinium nanocolloids in hyperlipidemic rabbits. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:569-78. [PMID: 25652897 DOI: 10.1016/j.nano.2014.12.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 12/09/2014] [Accepted: 12/13/2014] [Indexed: 10/24/2022]
Abstract
A high r1 relaxivity manganese-gadolinium nanocolloid (αvβ3-MnOL-Gd NC) was developed and effectively detected atherosclerotic angiogenesis in rabbits fed cholesterol-rich diets for 12 months using a clinical MRI scanner (3T). 3D mapping of neovasculature signal intensity revealed the spatial coherence and intensity of plaque angiogenic expansion, which may, with other high risk MR bioindicators, help identify high-risk patients with moderate (40% to 60%) vascular stenosis. Microscopy confirmed the predominant media and plaque distribution of fluorescent αvβ3-MnOL-Gd NC, mirroring the MR data. An expected close spatial association of αvβ3-integrin neovasculature and macrophages was noted, particularly within plaque shoulder regions. Manganese oleate bioelimination occurred via the biliary system into feces. Gd-DOTA was eliminated through the bile-fecal and renal excretion routes. αvβ3-MnOL-Gd NC offers an effective vehicle for T1w neovascular imaging in atherosclerosis. From the clinical editor: Cerebrovascular accidents are a leading cause of mortality and morbidity worldwide. The acute formation of thrombus following atherosclerotic plaque rupture has been well recognized as the etiology of stroke. The authors studied microanatomical features of vulnerable atherosclerotic plaque in this article, in an attempt to identify those with high risk of rupture. Gadolinium-manganese hybrid nanocolloid (MnOL-Gd NC) was developed as a novel contrast agent for MRI. They show that this agent is effective in providing neovascular imaging.
Collapse
Affiliation(s)
- Kezheng Wang
- Department of Radiology, the Fourth Hospital of Harbin Medical University and Molecular Imaging Center of Harbin Medical University, Harbin, China; Division of Cardiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Dipanjan Pan
- Division of Cardiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Anne H Schmieder
- Division of Cardiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Angana Senpan
- Division of Cardiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Shelton D Caruthers
- Division of Cardiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Grace Cui
- Division of Cardiology, Washington University School of Medicine, St. Louis, MO, USA
| | - John S Allen
- Division of Cardiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Huiying Zhang
- Division of Cardiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Baozhong Shen
- Department of Radiology, the Fourth Hospital of Harbin Medical University and Molecular Imaging Center of Harbin Medical University, Harbin, China.
| | - Gregory M Lanza
- Division of Cardiology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
106
|
Mulder WJM, Jaffer FA, Fayad ZA, Nahrendorf M. Imaging and nanomedicine in inflammatory atherosclerosis. Sci Transl Med 2015; 6:239sr1. [PMID: 24898749 DOI: 10.1126/scitranslmed.3005101] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Bioengineering provides unique opportunities to better understand and manage atherosclerotic disease. The field is entering a new era that merges the latest biological insights into inflammatory disease processes with targeted imaging and nanomedicine. Preclinical cardiovascular molecular imaging allows the in vivo study of targeted nanotherapeutics specifically directed toward immune system components that drive atherosclerotic plaque development and complication. The first multicenter trials highlight the potential contribution of multimodality imaging to more efficient drug development. This review describes how the integration of engineering, nanotechnology, and cardiovascular immunology may yield precision diagnostics and efficient therapeutics for atherosclerosis and its ischemic complications.
Collapse
Affiliation(s)
- Willem J M Mulder
- Translational and Molecular Imaging Institute, Department of Radiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA. Department of Vascular Medicine, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands.
| | - Farouc A Jaffer
- Cardiovascular Research Center and Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA
| | - Zahi A Fayad
- Translational and Molecular Imaging Institute, Department of Radiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
107
|
Maranhão RC, Tavares ER. Advances in non-invasive drug delivery for atherosclerotic heart disease. Expert Opin Drug Deliv 2015; 12:1135-47. [DOI: 10.1517/17425247.2015.999663] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
108
|
Li J, Dobrucki LW, Marjanovic M, Chaney EJ, Suslick KS, Boppart SA. Enhancement and wavelength-shifted emission of Cerenkov luminescence using multifunctional microspheres. Phys Med Biol 2015; 60:727-39. [PMID: 25555157 DOI: 10.1088/0031-9155/60/2/727] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cerenkov luminescence (CL) imaging is a new molecular imaging modality that utilizes the photons emitted during radioactive decay when charged particles travel faster than the phase velocity of light in a dielectric medium. Here we present a novel agent to convert and increase CL emission at longer wavelengths using multimodal protein microspheres (MSs). The (64)Cu-labeled protein microspheres contain quantum dots (QDs) encapsulated within a high-refractive-index-oil core. Dark box imaging of the MSs was conducted to demonstrate the improvement in CL emission at longer wavelengths. To illustrate the versatile design of these MSs and the potential of CL in disease diagnosis, these MSs were utilized for in vitro cell targeting and ex vivo CL-excited QD fluorescence (CL-FL) imaging of atherosclerotic plaques in rats. It was shown that by utilizing both QDs and MSs with a high-refractive-index-oil core, the CL emission increases by four-fold at longer wavelengths. Furthermore, we demonstrate that these MSs generate both an in vivo and ex vivo contrast signal. The design concept of utilizing QDs and high-index core MSs may contribute to future developments of in vivo CL imaging.
Collapse
Affiliation(s)
- Joanne Li
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA. Department of Nuclear, Plasma, and Radiological Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA. Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | | | | | | | | | | |
Collapse
|
109
|
PARK SJ, KIM JY, TEOH CL, KANG NY, CHANG YT. New Targets of Molecular Imaging in Atherosclerosis: Prehension of Current Status. ANAL SCI 2015; 31:245-55. [DOI: 10.2116/analsci.31.245] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Sung-Jin PARK
- Laboratory of Bioimaging Probe Development, Singapore Bioimaging Consortium, Agency for Science, Technology and Research
| | - Jun-Young KIM
- Laboratory of Bioimaging Probe Development, Singapore Bioimaging Consortium, Agency for Science, Technology and Research
| | - Chai Lean TEOH
- Laboratory of Bioimaging Probe Development, Singapore Bioimaging Consortium, Agency for Science, Technology and Research
| | - Nam-Young KANG
- Laboratory of Bioimaging Probe Development, Singapore Bioimaging Consortium, Agency for Science, Technology and Research
| | - Young-Tae CHANG
- Department of Chemistry & NUS MedChem Program of Life Sciences Institute, National University of Singapore
- Laboratory of Bioimaging Probe Development, Singapore Bioimaging Consortium, Agency for Science, Technology and Research
| |
Collapse
|
110
|
Xu X, Mao W, Chai Y, Dai J, Chen Q, Wang L, Zhuang Q, Pan Y, Chen M, Ni G, Huang Z. Angiogenesis Inhibitor, Endostar, Prevents Vasa Vasorum Neovascularization in a Swine Atherosclerosis Model. J Atheroscler Thromb 2015; 22:1100-12. [DOI: 10.5551/jat.26906] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Xiaoming Xu
- Department of Cardiology, Zhejiang Traditional Chinese Medical Hospital
| | - Wei Mao
- Department of Cardiology, Zhejiang Traditional Chinese Medical Hospital
| | - Yueyang Chai
- First College of Clinical Medicine, Zhejiang Chinese Medical University
| | - Jin Dai
- Department of Cardiology, Zhejiang Traditional Chinese Medical Hospital
| | - Qian Chen
- First College of Clinical Medicine, Zhejiang Chinese Medical University
| | - Lihui Wang
- First College of Clinical Medicine, Zhejiang Chinese Medical University
| | - Qin Zhuang
- First College of Clinical Medicine, Zhejiang Chinese Medical University
| | - Yongming Pan
- Center of Experimental Animals, Zhejiang Chinese Medical University
| | - Minli Chen
- Center of Experimental Animals, Zhejiang Chinese Medical University
| | - Guibao Ni
- Department of Pathology, Zhejiang Traditional Chinese Medical Hospital
| | - Zhaoquan Huang
- Department of Cardiology, Zhejiang Traditional Chinese Medical Hospital
| |
Collapse
|
111
|
Lavin B, Phinikaridou A, Henningsson M, Botnar RM. Current Development of Molecular Coronary Plaque Imaging using Magnetic Resonance Imaging towards Clinical Application. CURRENT CARDIOVASCULAR IMAGING REPORTS 2014. [DOI: 10.1007/s12410-014-9309-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
112
|
Iqbal B, Currie G, Greene L, Kiat H. Novel Radiopharmaceuticals in Cardiovascular Medicine: Present and Future. J Med Imaging Radiat Sci 2014; 45:423-434. [DOI: 10.1016/j.jmir.2014.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 09/03/2014] [Accepted: 09/05/2014] [Indexed: 01/25/2023]
|
113
|
Goette MJ, Lanza GM, Caruthers SD, Wickline SA. Improved quantitative (19) F MR molecular imaging with flip angle calibration and B1 -mapping compensation. J Magn Reson Imaging 2014; 42:488-94. [PMID: 25425244 DOI: 10.1002/jmri.24812] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 11/07/2014] [Indexed: 12/22/2022] Open
Abstract
PURPOSE To improve (19) F flip angle calibration and compensate for B1 inhomogeneities in quantitative (19) F MRI of sparse molecular epitopes with perfluorocarbon (PFC) nanoparticle (NP) emulsion contrast agents. MATERIALS AND METHODS Flip angle sweep experiments on PFC-NP point source phantoms with three custom-designed (19) F/(1) H dual-tuned coils revealed a difference in required power settings for (19) F and (1) H nuclei, which was used to calculate a calibration ratio specific for each coil. An image-based correction technique was developed using B1 -field mapping on (1) H to correct for (19) F and (1) H images in two phantom experiments. RESULTS Optimized (19) F peak power differed significantly from that of (1) H power for each coil (P < 0.05). A ratio of (19) F/(1) H power settings yielded a coil-specific and spatially independent calibration value (surface: 1.48 ± 0.06; semicylindrical: 1.71 ± 0.02, single-turn-solenoid: 1.92 ± 0.03). (1) H-image-based B1 correction equalized the signal intensity of (19) F images for two identical (19) F PFC-NP samples placed in different parts of the field, which were offset significantly by ~66% (P < 0.001), before correction. CONCLUSION (19) F flip angle calibration and B1 -mapping compensations to the (19) F images employing the more abundant (1) H signal as a basis for correction resulted in a significant change in the quantification of sparse (19) F MR signals from targeted PFC NP emulsions.
Collapse
Affiliation(s)
- Matthew J Goette
- Department of Biomedical Engineering, Washington University in St. Louis, Missouri, USA
| | - Gregory M Lanza
- Department of Biomedical Engineering, Washington University in St. Louis, Missouri, USA.,Department of Medicine, Washington University in St. Louis, Missouri, USA
| | - Shelton D Caruthers
- Department of Biomedical Engineering, Washington University in St. Louis, Missouri, USA.,Philips Healthcare, Cleveland, Ohio, USA
| | - Samuel A Wickline
- Department of Biomedical Engineering, Washington University in St. Louis, Missouri, USA.,Department of Medicine, Washington University in St. Louis, Missouri, USA
| |
Collapse
|
114
|
Sadat U, Jaffer FA, van Zandvoort MAMJ, Nicholls SJ, Ribatti D, Gillard JH. Inflammation and neovascularization intertwined in atherosclerosis: imaging of structural and molecular imaging targets. Circulation 2014; 130:786-94. [PMID: 25156914 DOI: 10.1161/circulationaha.114.010369] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Umar Sadat
- From the Cambridge Vascular Unit (U.S.) and University Department of Radiology (U.S., J.H.G.), Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, United Kingdom; Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, MA (F.A.J.); Advanced Microscopy Unit, Department of Genetics and Cell Biology-Molecular Cell Biology, Maastricht University, Maastricht, The Netherlands (M.A.M.J.v.Z.); Institute for Molecular Cardiovascular Research, Aachen University, Aachen, Germany (M.A.M.J.v.Z.); South Australian Health and Medical Research Institute and Heart Foundation Heart Health, University of Adelaide and Royal Adelaide Hospital, Adelaide, South Australia, Australia (S.J.N.); Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy (D.R.); and National Cancer Institute "Giovanni Paolo II," Bari, Italy (D.R.).
| | - Farouc A Jaffer
- From the Cambridge Vascular Unit (U.S.) and University Department of Radiology (U.S., J.H.G.), Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, United Kingdom; Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, MA (F.A.J.); Advanced Microscopy Unit, Department of Genetics and Cell Biology-Molecular Cell Biology, Maastricht University, Maastricht, The Netherlands (M.A.M.J.v.Z.); Institute for Molecular Cardiovascular Research, Aachen University, Aachen, Germany (M.A.M.J.v.Z.); South Australian Health and Medical Research Institute and Heart Foundation Heart Health, University of Adelaide and Royal Adelaide Hospital, Adelaide, South Australia, Australia (S.J.N.); Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy (D.R.); and National Cancer Institute "Giovanni Paolo II," Bari, Italy (D.R.)
| | - Marc A M J van Zandvoort
- From the Cambridge Vascular Unit (U.S.) and University Department of Radiology (U.S., J.H.G.), Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, United Kingdom; Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, MA (F.A.J.); Advanced Microscopy Unit, Department of Genetics and Cell Biology-Molecular Cell Biology, Maastricht University, Maastricht, The Netherlands (M.A.M.J.v.Z.); Institute for Molecular Cardiovascular Research, Aachen University, Aachen, Germany (M.A.M.J.v.Z.); South Australian Health and Medical Research Institute and Heart Foundation Heart Health, University of Adelaide and Royal Adelaide Hospital, Adelaide, South Australia, Australia (S.J.N.); Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy (D.R.); and National Cancer Institute "Giovanni Paolo II," Bari, Italy (D.R.)
| | - Stephen J Nicholls
- From the Cambridge Vascular Unit (U.S.) and University Department of Radiology (U.S., J.H.G.), Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, United Kingdom; Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, MA (F.A.J.); Advanced Microscopy Unit, Department of Genetics and Cell Biology-Molecular Cell Biology, Maastricht University, Maastricht, The Netherlands (M.A.M.J.v.Z.); Institute for Molecular Cardiovascular Research, Aachen University, Aachen, Germany (M.A.M.J.v.Z.); South Australian Health and Medical Research Institute and Heart Foundation Heart Health, University of Adelaide and Royal Adelaide Hospital, Adelaide, South Australia, Australia (S.J.N.); Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy (D.R.); and National Cancer Institute "Giovanni Paolo II," Bari, Italy (D.R.)
| | - Domenico Ribatti
- From the Cambridge Vascular Unit (U.S.) and University Department of Radiology (U.S., J.H.G.), Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, United Kingdom; Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, MA (F.A.J.); Advanced Microscopy Unit, Department of Genetics and Cell Biology-Molecular Cell Biology, Maastricht University, Maastricht, The Netherlands (M.A.M.J.v.Z.); Institute for Molecular Cardiovascular Research, Aachen University, Aachen, Germany (M.A.M.J.v.Z.); South Australian Health and Medical Research Institute and Heart Foundation Heart Health, University of Adelaide and Royal Adelaide Hospital, Adelaide, South Australia, Australia (S.J.N.); Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy (D.R.); and National Cancer Institute "Giovanni Paolo II," Bari, Italy (D.R.)
| | - Jonathan H Gillard
- From the Cambridge Vascular Unit (U.S.) and University Department of Radiology (U.S., J.H.G.), Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, United Kingdom; Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, MA (F.A.J.); Advanced Microscopy Unit, Department of Genetics and Cell Biology-Molecular Cell Biology, Maastricht University, Maastricht, The Netherlands (M.A.M.J.v.Z.); Institute for Molecular Cardiovascular Research, Aachen University, Aachen, Germany (M.A.M.J.v.Z.); South Australian Health and Medical Research Institute and Heart Foundation Heart Health, University of Adelaide and Royal Adelaide Hospital, Adelaide, South Australia, Australia (S.J.N.); Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy (D.R.); and National Cancer Institute "Giovanni Paolo II," Bari, Italy (D.R.)
| |
Collapse
|
115
|
Molecular imaging of plaques in coronary arteries with PET and SPECT. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2014; 11:259-73. [PMID: 25278976 PMCID: PMC4178519 DOI: 10.11909/j.issn.1671-5411.2014.03.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 08/15/2014] [Accepted: 08/19/2014] [Indexed: 01/26/2023]
Abstract
Coronary artery disease remains a major cause of mortality. Presence of atherosclerotic plaques in the coronary artery is responsible for lumen stenosis which is often used as an indicator for determining the severity of coronary artery disease. However, the degree of coronary lumen stenosis is not often related to compromising myocardial blood flow, as most of the cardiac events that are caused by atherosclerotic plaques are the result of vulnerable plaques which are prone to rupture. Thus, identification of vulnerable plaques in coronary arteries has become increasingly important to assist identify patients with high cardiovascular risks. Molecular imaging with use of positron emission tomography (PET) and single photon emission computed tomography (SPECT) has fulfilled this goal by providing functional information about plaque activity which enables accurate assessment of plaque stability. This review article provides an overview of diagnostic applications of molecular imaging techniques in the detection of plaques in coronary arteries with PET and SPECT. New radiopharmaceuticals used in the molecular imaging of coronary plaques and diagnostic applications of integrated PET/CT and PET/MRI in coronary plaques are also discussed.
Collapse
|
116
|
Fernández-Friera L, Ibáñez B, Fuster V. Imaging subclinical atherosclerosis: is it ready for prime time? A review. J Cardiovasc Transl Res 2014; 7:623-34. [PMID: 25119855 DOI: 10.1007/s12265-014-9582-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 07/28/2014] [Indexed: 01/02/2023]
Abstract
Imaging subclinical atherosclerosis holds the promise of individualized cardiovascular (CV) risk assessment. The large arsenal of noninvasive imaging techniques available today is playing an increasingly important role in the diagnosis and monitoring of subclinical atherosclerosis. However, there is a debate about the advisability of clinical screens for subclinical atherosclerosis and which modality is the most appropriate for monitoring risk and atherosclerosis progression. This article offers an overview of the traditional and emerging noninvasive imaging modalities used to detect early atherosclerosis, surveys population studies addressing the value of subclinical atherosclerosis detection, and also examines guideline recommendations for their clinical implementation. The clinical relevance of this manuscript lies in the potential of current imaging technology to improve CV risk prediction based on traditional risk factors and the present recommendations for subclinical atherosclerosis assessment. Noninvasive imaging will also help to identify individuals at high CV who would benefit from intensive prevention or therapeutic interventions.
Collapse
|
117
|
Iwaki S, Hokamura K, Ogawa M, Takehara Y, Muramatsu Y, Yamane T, Hirabayashi K, Morimoto Y, Hagisawa K, Nakahara K, Mineno T, Terai T, Komatsu T, Ueno T, Tamura K, Adachi Y, Hirata Y, Arita M, Arai H, Umemura K, Nagano T, Hanaoka K. A design strategy for small molecule-based targeted MRI contrast agents: their application for detection of atherosclerotic plaques. Org Biomol Chem 2014; 12:8611-8. [DOI: 10.1039/c4ob01270d] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
118
|
Winter P. Molecular Imaging at Nanoscale with Magnetic Resonance Imaging. Nanomedicine (Lond) 2014. [DOI: 10.1201/b17246-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
119
|
Khisamutdinov EF, Li H, Jasinski DL, Chen J, Fu J, Guo P. Enhancing immunomodulation on innate immunity by shape transition among RNA triangle, square and pentagon nanovehicles. Nucleic Acids Res 2014; 42:9996-10004. [PMID: 25092921 PMCID: PMC4150753 DOI: 10.1093/nar/gku516] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Modulation of immune response is important in cancer immunotherapy, vaccine adjuvant development and inflammatory or immune disease therapy. Here we report the development of new immunomodulators via control of shape transition among RNA triangle, square and pentagon. Changing one RNA strand in polygons automatically induced the stretching of the interior angle from 60° to 90° or 108°, resulting in self-assembly of elegant RNA triangles, squares and pentagons. When immunological adjuvants were incorporated, their immunomodulation effect for cytokine TNF-α and IL-6 induction was greatly enhanced in vitro and in animals up to 100-fold, while RNA polygon controls induced unnoticeable effect. The RNA nanoparticles were delivered to macrophages specifically. The degree of immunostimulation greatly depended on the size, shape and number of the payload per nanoparticles. Stronger immune response was observed when the number of adjuvants per polygon was increased, demonstrating the advantage of shape transition from triangle to pentagon.
Collapse
Affiliation(s)
- Emil F Khisamutdinov
- Department of Pharmaceutical Sciences, College of Pharmacy, Markey Cancer Center, Nanobiotechnology Center, University of Kentucky, Lexington, KY 40536, USA
| | - Hui Li
- Department of Pharmaceutical Sciences, College of Pharmacy, Markey Cancer Center, Nanobiotechnology Center, University of Kentucky, Lexington, KY 40536, USA
| | - Daniel L Jasinski
- Department of Pharmaceutical Sciences, College of Pharmacy, Markey Cancer Center, Nanobiotechnology Center, University of Kentucky, Lexington, KY 40536, USA
| | - Jiao Chen
- Center for Research on Environmental Disease, Graduate Center for Toxicology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Jian Fu
- Center for Research on Environmental Disease, Graduate Center for Toxicology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Peixuan Guo
- Department of Pharmaceutical Sciences, College of Pharmacy, Markey Cancer Center, Nanobiotechnology Center, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
120
|
Tsiapa I, Efthimiadou EK, Fragogeorgi E, Loudos G, Varvarigou AD, Bouziotis P, Kordas GC, Mihailidis D, Nikiforidis GC, Xanthopoulos S, Psimadas D, Paravatou-Petsotas M, Palamaris L, Hazle JD, Kagadis GC. (99m)Tc-labeled aminosilane-coated iron oxide nanoparticles for molecular imaging of ανβ3-mediated tumor expression and feasibility for hyperthermia treatment. J Colloid Interface Sci 2014; 433:163-175. [PMID: 25128864 DOI: 10.1016/j.jcis.2014.07.032] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 07/22/2014] [Accepted: 07/23/2014] [Indexed: 12/18/2022]
Abstract
HYPOTHESIS Dual-modality imaging agents, such as radiolabeled iron oxide nanoparticles (IO-NPs), are promising candidates for cancer diagnosis and therapy. We developed and evaluated aminosilane coated Fe3O4 (10±2nm) as a tumor imaging agent in nuclear medicine through 3-aminopropyltriethoxysilane (APTES) functionalization. We evaluated this multimeric system of targeted (99m)Tc-labeled nanoparticles (NPs) conjugated with a new RGD derivate (cRGDfK-Orn3-CGG), characterized as NPs-RGD as a potential thermal therapy delivery vehicle. EXPERIMENTS Transmission Electron Microscopy (TEM) and spectroscopy techniques were used to characterize the IO-NPs indicating their functionalization with peptides. Radiolabeled IO-NPs (targeted, non-targeted) were evaluated with regard to their radiochemical, radiobiological and imaging characteristics. In vivo studies were performed in normal and ανβ3-positive tumor (U87MG glioblastoma) bearing mice. We also demonstrated that this system could reach ablative temperatures in vivo. FINDINGS Both radiolabeled IO-NPs were obtained in high radiochemical yield (>98%) and proved stable in vitro. The in vivo studies for both IO-NPs have shown significant liver and spleen uptake at all examined time points in normal and U87MG glioblastoma tumor-bearing mice, due to their colloidal nature. We have confirmed through in vivo biodistribution studies that the non-targeted (99m)Tc-NPs poorly internalized in the tumor, while the targeted (99m)Tc-NPs-RGD, present 9-fold higher tumor accumulation at 1h p.i. Accumulation of both IO-NPs in other organs was negligible. Blocking experiments indicated target specificity for integrin receptors in U87MG glioblastoma cells. The preliminary in vivo study of applied alternating magnetic field showed that the induced hyperthermia is feasible due to the aid of IO-NPs.
Collapse
Affiliation(s)
- Irene Tsiapa
- Department of Medical Physics, School of Medicine, University of Patras, Patra, Greece; Institute for Nuclear and Radiological Sciences, Energy, Technology and Safety, National Center of Scientific Research "Demokritos", Aghia Paraskevi-Athens, Greece
| | - Eleni K Efthimiadou
- Sol-Gel Laboratory, Institute for Advanced Materials, Physicochemical Processes, Nanotechnology & Microsystems, NCSR "Demokritos", Aghia Paraskevi-Athens, Greece
| | - Eirini Fragogeorgi
- Department of Biomedical Technology Engineering, TEI of Athens, Aigaleo-Athens, Greece; Institute for Nuclear and Radiological Sciences, Energy, Technology and Safety, National Center of Scientific Research "Demokritos", Aghia Paraskevi-Athens, Greece
| | - George Loudos
- Department of Biomedical Technology Engineering, TEI of Athens, Aigaleo-Athens, Greece
| | - Alexandra D Varvarigou
- Institute for Nuclear and Radiological Sciences, Energy, Technology and Safety, National Center of Scientific Research "Demokritos", Aghia Paraskevi-Athens, Greece
| | - Penelope Bouziotis
- Institute for Nuclear and Radiological Sciences, Energy, Technology and Safety, National Center of Scientific Research "Demokritos", Aghia Paraskevi-Athens, Greece
| | - George C Kordas
- Department of Medical Physics, School of Medicine, University of Patras, Patra, Greece
| | | | - George C Nikiforidis
- Department of Medical Physics, School of Medicine, University of Patras, Patra, Greece
| | - Stavros Xanthopoulos
- Institute for Nuclear and Radiological Sciences, Energy, Technology and Safety, National Center of Scientific Research "Demokritos", Aghia Paraskevi-Athens, Greece
| | - Dimitrios Psimadas
- Department of Biomedical Technology Engineering, TEI of Athens, Aigaleo-Athens, Greece; Institute for Nuclear and Radiological Sciences, Energy, Technology and Safety, National Center of Scientific Research "Demokritos", Aghia Paraskevi-Athens, Greece
| | - Maria Paravatou-Petsotas
- Institute for Nuclear and Radiological Sciences, Energy, Technology and Safety, National Center of Scientific Research "Demokritos", Aghia Paraskevi-Athens, Greece
| | - Lazaros Palamaris
- Department of Biomedical Technology Engineering, TEI of Athens, Aigaleo-Athens, Greece
| | - John D Hazle
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - George C Kagadis
- Department of Medical Physics, School of Medicine, University of Patras, Patra, Greece; Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
121
|
Bala G, Cosyns B. Recent Advances in Visualizing Vulnerable Plaque: Focus on Noninvasive Molecular Imaging. Curr Cardiol Rep 2014; 16:520. [DOI: 10.1007/s11886-014-0520-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
122
|
Heo D, Lee E, Ku M, Hwang S, Kim B, Park Y, Han Lee Y, Huh YM, Haam S, Cheong JH, Yang J, Suh JS. Maleimidyl magnetic nanoplatform for facile molecular MRI. NANOTECHNOLOGY 2014; 25:275102. [PMID: 24960226 DOI: 10.1088/0957-4484/25/27/275102] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
In this study, we developed the maleimidyl magnetic nanoplatform, which enables functional targeting of a biomarker-specific moiety for molecular imaging via MRI. The maleimide group of the maleimidyl magnetic nanoplatform is conjugated with a thiol group without additional crosslinkers and side products. A physicochemical analysis was conducted to verify the effectiveness of the maleimidyl magnetic nanoplatform, and the existence of the maleimidyl group was investigated using the platform. To prepare biomarker-specific MRI probes, a thiolated aptamer and peptide were immobilized onto the maleimidyl group of the maleimidyl magnetic nanoplatform. The fabricated MRI probes were applied to four cancer cell lines: HT1080, MCF7, MKN45, and HEK293T. To investigate the potential of the molecular MRI probe, the target-biomarker specificity was confirmed without serious cytotoxicity, and in vivo MRI analysis using a xenograft mouse model was demonstrated. We believe these results will be useful for fabricating molecular MRI probes for the diagnosis of cancer.
Collapse
Affiliation(s)
- Dan Heo
- Department of Radiology, Yonsei University College of Medicine, Seoul, 120-752, Republic of Korea. Nanomedical National Core Research Center, Yonsei University, Seoul, 120-749, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
123
|
Shim CY, Lindner JR. Cardiovascular molecular imaging with contrast ultrasound: principles and applications. Korean Circ J 2014; 44:1-9. [PMID: 24497883 PMCID: PMC3905109 DOI: 10.4070/kcj.2014.44.1.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Methods for imaging the molecular or cellular profile of tissue are being developed for all forms of non-invasive cardiovascular imaging. It is thought that these technologies will potentially improve patient outcomes by allowing diagnosis of disease at an early-stage, monitoring disease progression, providing important information on patient risk, and for tailoring therapy to the molecular basis of disease. Molecular imaging is also already assuming an important role in science by providing a better understanding of the molecular basis of cardiovascular pathology, for assessing response to new therapies, and for rapidly optimizing new or established therapies. Ultrasound-based molecular imaging is one of these new approaches. Contrast-enhanced ultrasound molecular imaging relies on the detection of novel site-targeted microbubbles (MB) or other acoustically active particles which are administered by intravenous injection, circulate throughout the vascular compartment, and are then retained and imaged within regions of disease by ligand-directed binding. The technique is thought to be advantageous in practical terms of cost, time, and ease of use. The aim of this review is to discuss the molecular participants of cardiovascular disease that have been targeted for ultrasound imaging, general features of site-targeted MB, imaging protocols, and potential roles of ultrasound molecular imaging in cardiovascular research and clinical medicine.
Collapse
Affiliation(s)
- Chi Young Shim
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| | - Jonathan R Lindner
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
124
|
Fan Z, Sun Y, Di Chen, Tay D, Chen W, Deng CX, Fu J. Acoustic tweezing cytometry for live-cell subcellular modulation of intracellular cytoskeleton contractility. Sci Rep 2014; 3:2176. [PMID: 23846290 PMCID: PMC3709169 DOI: 10.1038/srep02176] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 06/24/2013] [Indexed: 11/09/2022] Open
Abstract
Mechanical forces are critical to modulate cell spreading, contractility, gene expression, and even stem cell differentiation. Yet, existing tools that can apply controllable subcellular forces to a large number of single cells simultaneously are still limited. Here we report a novel ultrasound tweezing cytometry utilizing ultrasound pulses to actuate functionalized lipid microbubbles covalently attached to single live cells to exert mechanical forces in the pN - nN range. Ultrasonic excitation of microbubbles could elicit a rapid and sustained reactive intracellular cytoskeleton contractile force increase in different adherent mechanosensitive cells. Further, ultrasound-mediated intracellular cytoskeleton contractility enhancement was dose-dependent and required an intact actin cytoskeleton as well as RhoA/ROCK signaling. Our results demonstrated the great potential of ultrasound tweezing cytometry technique using functionalized microbubbles as an actuatable, biocompatible, and multifunctional agent for biomechanical stimulations of cells.
Collapse
Affiliation(s)
- Zhenzhen Fan
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | | | | | | |
Collapse
|
125
|
Winter PM. Perfluorocarbon nanoparticles: evolution of a multimodality and multifunctional imaging agent. SCIENTIFICA 2014; 2014:746574. [PMID: 25024867 PMCID: PMC4082945 DOI: 10.1155/2014/746574] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2013] [Accepted: 05/20/2014] [Indexed: 06/03/2023]
Abstract
Perfluorocarbon nanoparticles offer a biologically inert, highly stable, and nontoxic platform that can be specifically designed to accomplish a range of molecular imaging and drug delivery functions in vivo. The particle surface can be decorated with targeting ligands to direct the agent to a variety of biomarkers that are associated with diseases such as cancer, cardiovascular disease, obesity, and thrombosis. The surface can also carry a high payload of imaging agents, ranging from paramagnetic metals for MRI, radionuclides for nuclear imaging, iodine for CT, and florescent tags for histology, allowing high sensitivity mapping of cellular receptors that may be expressed at very low levels in the body. In addition to these diagnostic imaging applications, the particles can be engineered to carry highly potent drugs and specifically deposit them into cell populations that display biosignatures of a variety of diseases. The highly flexible and robust nature of this combined molecular imaging and drug delivery vehicle has been exploited in a variety of animal models to demonstrate its potential impact on the care and treatment of patients suffering from some of the most debilitating diseases.
Collapse
Affiliation(s)
- Patrick M. Winter
- Department of Radiology, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA
| |
Collapse
|
126
|
Su H, Gorodny N, Gomez LF, Gangadharmath UB, Mu F, Chen G, Walsh JC, Szardenings K, Berman DS, Kolb HC, Tamarappoo BK. Atherosclerotic plaque uptake of a novel integrin tracer ¹⁸F-Flotegatide in a mouse model of atherosclerosis. J Nucl Cardiol 2014; 21:553-62. [PMID: 24627345 PMCID: PMC4316660 DOI: 10.1007/s12350-014-9879-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 02/11/2014] [Indexed: 10/25/2022]
Abstract
INTRODUCTION Rupture of unstable atherosclerotic plaque that leads to stroke and myocardial infarction may be induced by macrophage infiltration and neovessel formation. A tracer that selectively binds to integrin αvβ3 a protein expressed by macrophages and neovascular endothelium may identify rupture prone plaque. METHODS (18)F-labeled "R-G-D" containing tripeptide (Flotegatide), a click chemistry derived radiotracer that binds to integrin αvβ3 was injected in ApoE knockout mice fed a high fat diet. Uptake of Flotegatide by atherosclerotic plaque was visualized by micro-PET, autoradiography, and correlated to histologic markers of inflammation and angiogenesis. RESULTS We found that Flotegatide preferentially binds to aortic plaque in an ApoE knockout mouse model of atherosclerosis. The tracer's uptake is strongly associated with presence of histologic markers for macrophage infiltration and integrin expression. There is a weaker but detectable association between Flotegatide uptake and presence of an immunohistochemical marker for neovascularization. DISCUSSION We hypothesize that Flotegatide may be a useful tracer for visualization of inflamed plaque in clinical subjects with atherosclerosis and may have potential for detecting vulnerable plaque.
Collapse
Affiliation(s)
- Helen Su
- Siemens Molecular Imaging, 6140 Bristol Parkway, Culver City, CA, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
127
|
MR cholangiography demonstrates unsuspected rapid biliary clearance of nanoparticles in rodents: implications for clinical translation. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2014; 10:1385-8. [PMID: 24832959 DOI: 10.1016/j.nano.2014.05.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 05/02/2014] [Accepted: 05/02/2014] [Indexed: 02/02/2023]
Abstract
Due to their small size, lower cost, short reproduction cycle, and genetic manipulation, rodents have been widely used to test the safety and efficacy for pharmaceutical development in human disease. In this report, MR cholangiography demonstrated an unexpected rapid (<5 min) biliary elimination of gadolinium-perfluorocarbon nanoparticles (approximately 250 nm diameter) into the common bile duct and small intestine of rats, which is notably different from nanoparticle clearance patterns in larger animals and humans. Unawareness of this dissimilarity in nanoparticle clearance mechanisms between small animals and humans may lead to fundamental errors in predicting nanoparticle efficacy, pharmacokinetics, biodistribution, bioelimination, and toxicity. From the clinical editor: Comprehensive understanding of nanoparticle clearance is a clear prerequisite for human applications of nanomedicine-based therapeutic approaches. Through a novel use of MR cholangiography, this study demonstrates unusually rapid hepatic clearance of gadolinium-perfluorocarbon nanoparticles in rodents, in a pattern that is different than what is observed in larger animals and humans, raising awareness of important differences between common rodent-based models and larger mammals.
Collapse
|
128
|
Phinikaridou A, Andia ME, Indermuehle A, Onthank DC, Cesati RR, Smith A, Robinson SP, Saha P, Botnar RM. Vascular Remodeling and Plaque Vulnerability in a Rabbit Model of Atherosclerosis: Comparison of Delayed-Enhancement MR Imaging with an Elastin-specific Contrast Agent and Unenhanced Black-Blood MR Imaging. Radiology 2014; 271:390-9. [DOI: 10.1148/radiol.13130502] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
129
|
Zhang Y, Guallar E, Qiao Y, Wasserman BA. Is carotid intima-media thickness as predictive as other noninvasive techniques for the detection of coronary artery disease? Arterioscler Thromb Vasc Biol 2014; 34:1341-5. [PMID: 24764454 DOI: 10.1161/atvbaha.113.302075] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Carotid intima-media thickness (CIMT) measured by B-mode ultrasound is the most widely used noninvasive imaging method to assess atherosclerosis and cardiovascular risk. CIMT has been consistently associated with coronary artery disease and stroke; however, recent meta-analyses and systematic reviews suggest that its clinical usefulness may be limited because the addition of CIMT to traditional risk factors has not improved the risk prediction of cardiovascular events in the general population. Characterizing the carotid wall by MRI may have greater clinical utility compared with CIMT measurements by ultrasound. Unlike CIMT, MRI measurements of wall thickness include the adventitia and may be sensitive to adventitial thickening that results from vasa vasorum proliferation as a sign of early plaque development. MRI also has the ability to image the entire circumference of the carotid wall, including the outer wall of the carotid bulb where plaque forms in its earliest stage, and identify plaque components such as the lipid core, fibrous cap, and intraplaque hemorrhage that are closely related to plaque vulnerability and cardiovascular risk. Additional research is needed to assess the added prognostic value of MRI measurements of wall and plaque features in risk prediction beyond traditional risk factors.
Collapse
Affiliation(s)
- Yiyi Zhang
- From the Departments of Epidemiology and Medicine, and Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD (Y.Z., E.G.); and Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins Hospital, Baltimore, MD (Y.Q., B.A.W.)
| | - Eliseo Guallar
- From the Departments of Epidemiology and Medicine, and Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD (Y.Z., E.G.); and Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins Hospital, Baltimore, MD (Y.Q., B.A.W.)
| | - Ye Qiao
- From the Departments of Epidemiology and Medicine, and Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD (Y.Z., E.G.); and Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins Hospital, Baltimore, MD (Y.Q., B.A.W.)
| | - Bruce A Wasserman
- From the Departments of Epidemiology and Medicine, and Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD (Y.Z., E.G.); and Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins Hospital, Baltimore, MD (Y.Q., B.A.W.).
| |
Collapse
|
130
|
Jallouk AP, Moley KH, Omurtag K, Hu G, Lanza GM, Wickline SA, Hood JL. Nanoparticle incorporation of melittin reduces sperm and vaginal epithelium cytotoxicity. PLoS One 2014; 9:e95411. [PMID: 24748389 PMCID: PMC3991669 DOI: 10.1371/journal.pone.0095411] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 03/27/2014] [Indexed: 11/18/2022] Open
Abstract
Melittin is a cytolytic peptide component of bee venom which rapidly integrates into lipid bilayers and forms pores resulting in osmotic lysis. While the therapeutic utility of free melittin is limited by its cytotoxicity, incorporation of melittin into the lipid shell of a perfluorocarbon nanoparticle has been shown to reduce its toxicity in vivo. Our group has previously demonstrated that perfluorocarbon nanoparticles containing melittin at concentrations <10 µM inhibit HIV infectivity in vitro. In the current study, we assessed the impact of blank and melittin-containing perfluorocarbon nanoparticles on sperm motility and the viability of both sperm and vaginal epithelial cells. We found that free melittin was toxic to sperm and vaginal epithelium at concentrations greater than 2 µM (p<0.001). However, melittin nanoparticles were not cytotoxic to sperm (p = 0.42) or vaginal epithelium (p = 0.48) at an equivalent melittin concentration of 10 µM. Thus, nanoparticle formulation of melittin reduced melittin cytotoxicity fivefold and prevented melittin toxicity at concentrations previously shown to inhibit HIV infectivity. Melittin nanoparticles were toxic to vaginal epithelium at equivalent melittin concentrations ≥20 µM (p<0.001) and were toxic to sperm at equivalent melittin concentrations ≥40 µM (p<0.001). Sperm cytotoxicity was enhanced by targeting of the nanoparticles to the sperm surface antigen sperm adhesion molecule 1. While further testing is needed to determine the extent of cytotoxicity in a more physiologically relevant model system, these results suggest that melittin-containing nanoparticles could form the basis of a virucide that is not toxic to sperm and vaginal epithelium. This virucide would be beneficial for HIV serodiscordant couples seeking to achieve natural pregnancy.
Collapse
Affiliation(s)
- Andrew P. Jallouk
- Department of Medicine, Division of Cardiology, Consortium for Translational Research in Advanced Imaging and Nanomedicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Kelle H. Moley
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Kenan Omurtag
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Grace Hu
- Department of Medicine, Division of Cardiology, Consortium for Translational Research in Advanced Imaging and Nanomedicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Gregory M. Lanza
- Department of Medicine, Division of Cardiology, Consortium for Translational Research in Advanced Imaging and Nanomedicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Samuel A. Wickline
- Department of Medicine, Division of Cardiology, Consortium for Translational Research in Advanced Imaging and Nanomedicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Joshua L. Hood
- Department of Medicine, Division of Cardiology, Consortium for Translational Research in Advanced Imaging and Nanomedicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
131
|
Wen S, Liu DF, Cui Y, Harris SS, Chen YC, Li KC, Ju SH, Teng GJ. In vivo MRI detection of carotid atherosclerotic lesions and kidney inflammation in ApoE-deficient mice by using LOX-1 targeted iron nanoparticles. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2014; 10:639-49. [DOI: 10.1016/j.nano.2013.09.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 08/12/2013] [Accepted: 09/24/2013] [Indexed: 12/18/2022]
|
132
|
Xue S, Qiao J, Jiang J, Hubbard K, White N, Wei L, Li S, Liu ZR, Yang JJ. Design of ProCAs (protein-based Gd(3+) MRI contrast agents) with high dose efficiency and capability for molecular imaging of cancer biomarkers. Med Res Rev 2014; 34:1070-99. [PMID: 24615853 DOI: 10.1002/med.21313] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Magnetic resonance imaging (MRI) is the leading imaging technique for disease diagnostics, providing high resolution, three-dimensional images noninvasively. MRI contrast agents are designed to improve the contrast and sensitivity of MRI. However, current clinically used MRI contrast agents have relaxivities far below the theoretical upper limit, which largely prevent advancing molecular imaging of biomarkers with desired sensitivity and specificity. This review describes current progress in the development of a new class of protein-based MRI contrast agents (ProCAs) with high relaxivity using protein design to optimize the parameters that govern relaxivity. Further, engineering with targeting moiety allows these contrast agents to be applicable for molecular imaging of prostate cancer biomarkers by MRI. The developed protein-based contrast agents also exhibit additional in vitro and in vivo advantages for molecular imaging of disease biomarkers, such as high metal-binding stability and selectivity, reduced toxicity, proper blood circulation time, and higher permeability in tumor tissue in addition to improved relaxivities.
Collapse
Affiliation(s)
- Shenghui Xue
- Departments of Chemistry and Biology, Georgia State University, Atlanta, Georgia; Center for Diagnostics & Therapeutics (CDT), Georgia State University, Atlanta, Georgia; Center for Biotechnology and Drug Design, Georgia State University, Atlanta, Georgia
| | | | | | | | | | | | | | | | | |
Collapse
|
133
|
The Great Migration: How MRI Replaces Traditional Imaging Techniques for the Characterization of Atherosclerosis. CURRENT RADIOLOGY REPORTS 2014. [DOI: 10.1007/s40134-013-0040-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
134
|
Silva Marques J, Pinto FJ. The vulnerable plaque: current concepts and future perspectives on coronary morphology, composition and wall stress imaging. Rev Port Cardiol 2014; 33:101-10. [PMID: 24513090 DOI: 10.1016/j.repc.2013.07.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 07/22/2013] [Indexed: 12/17/2022] Open
Abstract
Cardiovascular imaging plays an important role in the identification and characterization of the vulnerable plaque. A major goal is the ability to identify individuals at risk of plaque rupture and developing an acute coronary syndrome. Early recognition of rupture-prone atherosclerotic plaques may lead to the development of pharmacologic and interventional strategies to reduce acute coronary events. We review state-of-the-art cardiovascular imaging for identification of the vulnerable plaque. There is ample evidence of a close relationship between plaque morphology and patient outcome, but molecular imaging can add significant information on tissue characterization, inflammation and subclinical thrombosis. Additionally, identifying arterial wall exposed to high shear stress may further identify rupture-prone arterial segments. These new modalities may help reduce the individual, social and economic burden of cardiovascular disease.
Collapse
Affiliation(s)
- João Silva Marques
- University Hospital Santa Maria, Department of Cardiology I, Lisbon Academic Medical Centre, CCUL, Lisbon, Portugal.
| | - Fausto J Pinto
- University Hospital Santa Maria, Department of Cardiology I, Lisbon Academic Medical Centre, CCUL, Lisbon, Portugal
| |
Collapse
|
135
|
The vulnerable plaque: Current concepts and future perspectives on coronary morphology, composition and wall stress imaging. REVISTA PORTUGUESA DE CARDIOLOGIA (ENGLISH EDITION) 2014. [DOI: 10.1016/j.repce.2013.07.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
136
|
Tong S, Fine EJ, Lin Y, Cradick TJ, Bao G. Nanomedicine: tiny particles and machines give huge gains. Ann Biomed Eng 2014; 42:243-59. [PMID: 24297494 PMCID: PMC3962788 DOI: 10.1007/s10439-013-0952-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 11/20/2013] [Indexed: 12/13/2022]
Abstract
Nanomedicine is an emerging field that integrates nanotechnology, biomolecular engineering, life sciences and medicine; it is expected to produce major breakthroughs in medical diagnostics and therapeutics. Nano-scale structures and devices are compatible in size with proteins and nucleic acids in living cells. Therefore, the design, characterization and application of nano-scale probes, carriers and machines may provide unprecedented opportunities for achieving a better control of biological processes, and drastic improvements in disease detection, therapy, and prevention. Recent advances in nanomedicine include the development of nanoparticle (NP)-based probes for molecular imaging, nano-carriers for drug/gene delivery, multifunctional NPs for theranostics, and molecular machines for biological and medical studies. This article provides an overview of the nanomedicine field, with an emphasis on NPs for imaging and therapy, as well as engineered nucleases for genome editing. The challenges in translating nanomedicine approaches to clinical applications are discussed.
Collapse
Affiliation(s)
- Sheng Tong
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Eli J. Fine
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Yanni Lin
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Thomas J. Cradick
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Gang Bao
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| |
Collapse
|
137
|
Weissleder R, Nahrendorf M, Pittet MJ. Imaging macrophages with nanoparticles. NATURE MATERIALS 2014; 13:125-38. [PMID: 24452356 DOI: 10.1038/nmat3780] [Citation(s) in RCA: 570] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2013] [Accepted: 09/17/2013] [Indexed: 05/02/2023]
Abstract
Nanomaterials have much to offer, not only in deciphering innate immune cell biology and tracking cells, but also in advancing personalized clinical care by providing diagnostic and prognostic information, quantifying treatment efficacy and designing better therapeutics. This Review presents different types of nanomaterial, their biological properties and their applications for imaging macrophages in human diseases, including cancer, atherosclerosis, myocardial infarction, aortic aneurysm, diabetes and other conditions. We anticipate that future needs will include the development of nanomaterials that are specific for immune cell subsets and can be used as imaging surrogates for nanotherapeutics. New in vivo imaging clinical tools for noninvasive macrophage quantification are thus ultimately expected to become relevant to predicting patients' clinical outcome, defining treatment options and monitoring responses to therapy.
Collapse
Affiliation(s)
- Ralph Weissleder
- 1] Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, USA [2] Department of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Boston, Massachusetts 02115, USA [3] Department of Radiology, Massachusetts General Hospital, 32 Fruit Street, Boston, Massachusetts 02114, USA
| | - Matthias Nahrendorf
- 1] Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, USA [2] Department of Radiology, Massachusetts General Hospital, 32 Fruit Street, Boston, Massachusetts 02114, USA
| | - Mikael J Pittet
- 1] Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, USA [2] Department of Radiology, Massachusetts General Hospital, 32 Fruit Street, Boston, Massachusetts 02114, USA
| |
Collapse
|
138
|
Xie S, Dong B, Sun X, Tala, He X, Zhou J, Liu M, Li D. Identification of a cytoplasmic linker protein as a potential target for neovascularization. Atherosclerosis 2014; 233:403-409. [PMID: 24530770 DOI: 10.1016/j.atherosclerosis.2014.01.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Revised: 12/02/2013] [Accepted: 01/02/2014] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Atherosclerosis and other cardiovascular diseases are serious threats to human health and become the leading cause of death in the world. Emerging evidence reveals that inhibition of plaque neovascularization could be an effective approach for the treatment of atherosclerosis. This study was conducted to identify cytoplasmic linker protein 170 as a potential target for cardiovascular diseases through modulation of neovascularization. METHODS AND RESULTS Immunofluorescence microscopy revealed that cytoplasmic linker protein 170 was ubiquitously expressed in mouse kidney, liver, lung, normal non-atherosclerotic aorta, and atherosclerotic aorta and was partly localized in the vascular endothelium. siRNAs were introduced to human umbilical vein endothelial cells (HUVECs) and the effect of knockdown was confirmed by Western blotting. Vascularization study was assessed with matrigel-based capillary assembly, branching, and in vivo matrigel plug assays. The data showed that siRNA-mediated knockdown of the cytoplasmic linker protein remarkably compromised the assembly and branching of capillary-like blood vessels and neovascularization in vivo. Cell motility and polarity properties were then analyzed using scratch wound repair, boyden chamber, and immunofluorescence assays, and the results revealed that the cytoplasmic linker protein was critical for the motility abilities of HUVECs through its actions on cell polarity. CONCLUSION Both in vitro and in vivo studies demonstrate the significance of the cytoplasmic linker protein for blood vessel formation. Mechanistic investigation reveals that its effect on neovascularization is orchestrated through its regulation of vascular endothelial cell polarity and motility. These findings provide the basis for exploring effective approaches to regulate neovascularization in cardiovascular diseases.
Collapse
Affiliation(s)
- Songbo Xie
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Bin Dong
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Xiaodong Sun
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Tala
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Xianfei He
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Jun Zhou
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Min Liu
- Department of Biochemistry, Basic Medical College, Tianjin Medical University, Tianjin 300070, China.
| | - Dengwen Li
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China.
| |
Collapse
|
139
|
Abstract
Nanoparticles are rapidly being developed and trialed to overcome several limitations of traditional drug delivery systems and are coming up as a distinct therapeutics for cancer treatment. Conventional chemotherapeutics possess some serious side effects including damage of the immune system and other organs with rapidly proliferating cells due to nonspecific targeting, lack of solubility, and inability to enter the core of the tumors resulting in impaired treatment with reduced dose and with low survival rate. Nanotechnology has provided the opportunity to get direct access of the cancerous cells selectively with increased drug localization and cellular uptake. Nanoparticles can be programmed for recognizing the cancerous cells and giving selective and accurate drug delivery avoiding interaction with the healthy cells. This review focuses on cell recognizing ability of nanoparticles by various strategies having unique identifying properties that distinguish them from previous anticancer therapies. It also discusses specific drug delivery by nanoparticles inside the cells illustrating many successful researches and how nanoparticles remove the side effects of conventional therapies with tailored cancer treatment.
Collapse
|
140
|
Zhuo Y, Hu H, Chen W, Lu M, Tian L, Yu H, Long KD, Chow E, King WP, Singamaneni S, Cunningham BT. Single nanoparticle detection using photonic crystal enhanced microscopy. Analyst 2014; 139:1007-15. [PMID: 24432353 DOI: 10.1039/c3an02295a] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
We demonstrate a label-free biosensor imaging approach that utilizes a photonic crystal (PC) surface to detect surface attachment of individual dielectric and metal nanoparticles through measurement of localized shifts in the resonant wavelength and resonant reflection magnitude from the PC. Using a microscopy-based approach to scan the PC resonant reflection properties with 0.6 μm spatial resolution, we show that metal nanoparticles attached to the biosensor surface with strong absorption at the resonant wavelength induce a highly localized reduction in reflection efficiency and are able to be detected by modulation of the resonant wavelength. Experimental demonstrations of single-nanoparticle imaging are supported by finite-difference time-domain computer simulations. The ability to image surface-adsorption of individual nanoparticles offers a route to single molecule biosensing, in which the particles can be functionalized with specific recognition molecules and utilized as tags.
Collapse
Affiliation(s)
- Yue Zhuo
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Cyrus T, Winter PM, Caruthers SD, Wickline SA, Lanza GM. Magnetic resonance nanoparticles for cardiovascular molecular imaging and therapy. Expert Rev Cardiovasc Ther 2014; 3:705-15. [PMID: 16076280 DOI: 10.1586/14779072.3.4.705] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Molecular vascular imaging represents a novel tool that promises to change the current medical paradigm of 'see and treat' to a 'detect and prevent' strategy. Nanoparticle agents, such as superparamagnetic nanoparticles and perfluorocarbon nanoparticle emulsions, have been developed for noninvasive imaging, particularly for magnetic resonance imaging. Designed to target specific epitopes in tissues, these agents are beginning to enter clinical trials for cardiovascular applications. The delivery of local therapy with these nanoparticles, using mechanisms such as contact-facilitated drug delivery, is in the advanced stages of preclinical research. Ultimately, combined diagnostic and therapeutic nanoparticle formulations may allow patients to be characterized noninvasively and segmented to receive custom-tailored therapy. This review focuses on recent developments of nanoparticle technologies with an emphasis on cardiovascular applications of magnetic resonance imaging.
Collapse
Affiliation(s)
- Tillmann Cyrus
- Washington University School of Medicine, Barnes-Jewish Hospital, 660 South Euclid Ave., Box 8086, Saint Louis, MO 63110, USA.
| | | | | | | | | |
Collapse
|
142
|
Beer AJ, Pelisek J, Heider P, Saraste A, Reeps C, Metz S, Seidl S, Kessler H, Wester HJ, Eckstein HH, Schwaiger M. PET/CT imaging of integrin αvβ3 expression in human carotid atherosclerosis. JACC Cardiovasc Imaging 2014; 7:178-87. [PMID: 24412187 DOI: 10.1016/j.jcmg.2013.12.003] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 12/05/2013] [Indexed: 12/12/2022]
Abstract
OBJECTIVES The goal of this study was to evaluate the feasibility of [(18)F]Galacto-RGD positron emission tomography (PET)/computed tomography (CT) imaging of αvβ3 expression in human carotid plaques. BACKGROUND The integrin αvβ3 is expressed by macrophages and angiogenic endothelial cells in atherosclerotic lesions and thus is a marker of plaque inflammation and, potentially, of plaque vulnerability. [(18)F]Galacto-RGD is a PET tracer binding specifically to αvβ3. Therefore, [(18)F]Galacto-RGD PET/CT imaging of αvβ3 expression in human carotid plaques might provide a novel noninvasive biomarker of plaque vulnerability. METHODS [(18)F]Galacto-RGD PET/CT imaging was performed in 10 patients with high-grade carotid artery stenosis scheduled for carotid endarterectomy. Tracer uptake was measured in the stenotic areas of the carotid arteries, as well as on the contralateral side, and was corrected for blood pool activity, measured in the distal common carotid artery (target-to-background [TB] ratio). TB ratio was correlated with immunohistochemistry of αvβ3 expression (LM609), macrophage density (CD68), and microvessel density (CD31) of the surgical specimen. In addition, ex vivo autoradiography of the surgical specimen with [(18)F]Galacto-RGD and competition experiments with an unlabeled αvβ3-specific RGD peptide were performed. RESULTS [(18)F]Galacto-RGD PET/CT showed significantly higher TB ratios in stenotic areas compared with nonstenotic areas (p = 0.01). TB ratios correlated significantly with αvβ3 expression (R = 0.787, p = 0.026) and intensity of ex vivo autoradiography (R = 0.733, p = 0.038). Binding to atherosclerotic plaques was efficiently blocked in ex vivo competition experiments. A weak-to-moderate correlation was found with macrophage density (R = 0.367, p = 0.299) and microvessel density (R = 0.479, p = 0.176), which did not reach statistical significance. CONCLUSIONS [(18)F]Galacto-RGD PET/CT shows specific tracer accumulation in human atherosclerotic carotid plaques, which correlates with αvβ3 expression. Based on these initial data, larger prospective studies are now warranted to evaluate the potential of molecular imaging of αvβ3 expression for assessment of plaque inflammation in patients.
Collapse
Affiliation(s)
- Ambros J Beer
- Department of Nuclear Medicine, Technische Universität München, Munich, Germany.
| | - Jaroslav Pelisek
- Department of Vascular Surgery, Technische Universität München, Munich, Germany
| | - Peter Heider
- Department of Vascular Surgery, Technische Universität München, Munich, Germany
| | - Antti Saraste
- Turku PET Centre and Department of Cardiology, Turku, Finland
| | - Christian Reeps
- Department of Vascular Surgery, Technische Universität München, Munich, Germany
| | - Stephan Metz
- Department of Radiology, Technische Universität München, Munich, Germany
| | - Stefan Seidl
- Department of Pathology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Horst Kessler
- Institute for Advanced Study and Center of Integrated Protein Science, Technische Universität München, Department Chemie, Garching, Germany; Chemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hans-Jürgen Wester
- Chair of Pharmaceutical Radiochemistry, Technische Universität München, Garching, Germany
| | | | - Markus Schwaiger
- Department of Nuclear Medicine, Technische Universität München, Munich, Germany
| |
Collapse
|
143
|
Abstract
Current advances in nanotechnology have paved the way for the early detection, prevention and treatment of various diseases such as vascular disorders and cancer. These advances have provided novel approaches or modalities of incorporating or adsorbing therapeutic, biosensor and targeting agents into/on nanoparticles. With significant progress, nanomedicine for vascular therapy has shown significant advantages over traditional medicine because of its ability to selectively target the disease site and reduce adverse side effects. Targeted delivery of nanoparticles to vascular endothelial cells or the vascular wall provides an effective and more efficient way for early detection and/or treatment of vascular diseases such as atherosclerosis, thrombosis and Cerebrovascular Amyloid Angiopathy (CAA). Clinical applications of biocompatible and biodegradable polymers in areas such as vascular graft, implantable drug delivery, stent devices and tissue engineering scaffolds have advanced the candidature of polymers as potential nano-carriers for vascular-targeted delivery of diagnostic agents and drugs. This review focuses on the basic aspects of the vasculature and its associated diseases and relates them to polymeric nanoparticle-based strategies for targeting therapeutic agents to diseased vascular site.
Collapse
Affiliation(s)
- Edward Agyare
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL ; Division of Radiation Oncology, University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Karunyna Kandimalla
- Department of Pharmaceutics and Brain Barriers Research Center, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
144
|
Abstract
Techniques for in vivo assessment of disease-related molecular changes are being developed for all forms of non-invasive cardiovascular imaging. The ability to evaluate tissue molecular or cellular phenotype in patients has the potential to not only improve diagnostic capabilities but to enhance clinical care either by detecting disease at an earlier stage when it is more amenable to therapy, or by guiding most appropriate therapies. These new techniques also can be used in research programs in order to characterize pathophysiology and as a surrogate endpoint for therapeutic efficacy. The most common approach for molecular imaging involves the creation of novel-targeted contrast agents that are designed so that their kinetic properties are different in disease tissues. The main focus of this review is not to describe all the different molecular imaging approaches that have been developed, but rather to describe the status of the field and highlight some of the clinical and research applications that molecular imaging will likely provide meaningful benefit. Specific target areas include assessment of atherosclerotic disease, tissue ischemia, and ventricular and vascular remodeling.
Collapse
Affiliation(s)
- Jonathan R Lindner
- Knight Cardiovascular Institute, Oregon Health & Science University, UHN-62, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA,
| | | |
Collapse
|
145
|
Makowski MR, Botnar RM. MR imaging of the arterial vessel wall: molecular imaging from bench to bedside. Radiology 2013; 269:34-51. [PMID: 24062561 DOI: 10.1148/radiol.13102336] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Cardiovascular diseases remain the leading cause of morbidity and mortality in the Western world and developing countries. In clinical practice, in vivo characterization of atherosclerotic lesions causing myocardial infarction, ischemic stroke, and other complications remains challenging. Imaging methods, limited to the assessment luminal stenosis, are the current reference standard for the assessment of clinically significant coronary and carotid artery disease and the guidance of treatment. These techniques do not allow distinction between stable and potentially vulnerable atherosclerotic plaque. Magnetic resonance (MR) imaging is a modality well suited for visualization and characterization of the relatively thin arterial vessel wall, because it allows imaging with high spatial resolution and excellent soft-tissue contrast. In clinical practice, atherosclerotic plaque components of the carotid artery and aorta may be differentiated and characterized by using unenhanced vessel wall MR imaging. Additional information can be gained by using clinically approved nonspecific contrast agents. With the advent of targeted MR contrast agents, which enhance specific molecules or cells, pathologic processes can be visualized at a molecular level with high spatial resolution. In this article, the pathophysiologic changes of the arterial vessel wall underlying the development of atherosclerosis will be first reviewed. Then basic principles and properties of molecular MR imaging contrast agents will be introduced. Additionally, recent advances in preclinical molecular vessel wall imaging will be reviewed. Finally, the clinical feasibility of arterial vessel wall imaging at unenhanced and contrast material-enhanced MR imaging of the aortic, carotid, and coronary vessel wall will be discussed.
Collapse
Affiliation(s)
- Marcus R Makowski
- Division of Imaging Sciences, BHF Centre of Excellence, Wellcome Trust and EPSRC Medical Engineering Center, and NIHR Biomedical Research Centre, King's College London, 4th Floor, Lambeth Wing, St Thomas Hospital, London SE1 7EH, England
| | | |
Collapse
|
146
|
Phinikaridou A, Andia ME, Lacerda S, Lorrio S, Makowski MR, Botnar RM. Molecular MRI of atherosclerosis. Molecules 2013; 18:14042-69. [PMID: 24232739 PMCID: PMC6270261 DOI: 10.3390/molecules181114042] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 10/29/2013] [Accepted: 10/29/2013] [Indexed: 11/22/2022] Open
Abstract
Despite advances in prevention, risk assessment and treatment, coronary artery disease (CAD) remains the leading cause of morbidity and mortality in Western countries. The lion's share is due to acute coronary syndromes (ACS), which are predominantly triggered by plaque rupture or erosion and subsequent coronary thrombosis. As the majority of vulnerable plaques does not cause a significant stenosis, due to expansive remodeling, and are rather defined by their composition and biological activity, detection of vulnerable plaques with x-ray angiography has shown little success. Non-invasive vulnerable plaque detection by identifying biological features that have been associated with plaque progression, destabilization and rupture may therefore be more appropriate and may allow earlier detection, more aggressive treatment and monitoring of treatment response. MR molecular imaging with target specific molecular probes has shown great promise for the noninvasive in vivo visualization of biological processes at the molecular and cellular level in animals and humans. Compared to other imaging modalities; MRI can provide excellent spatial resolution; high soft tissue contrast and has the ability to simultaneously image anatomy; function as well as biological tissue composition and activity.
Collapse
Affiliation(s)
- Alkystis Phinikaridou
- Division of Imaging Sciences and Biomedical Engineering, King’s College London, 4th Floor, Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, UK; E-Mails: (A.P.); (M.E.A.); (S.L.); (S.L.); (M.R.M.)
| | - Marcelo E. Andia
- Division of Imaging Sciences and Biomedical Engineering, King’s College London, 4th Floor, Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, UK; E-Mails: (A.P.); (M.E.A.); (S.L.); (S.L.); (M.R.M.)
- Radiology Department, School of Medicine, Pontificia Universidad Catolica de Chile, Santiago 8331150, Chile
| | - Sara Lacerda
- Division of Imaging Sciences and Biomedical Engineering, King’s College London, 4th Floor, Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, UK; E-Mails: (A.P.); (M.E.A.); (S.L.); (S.L.); (M.R.M.)
| | - Silvia Lorrio
- Division of Imaging Sciences and Biomedical Engineering, King’s College London, 4th Floor, Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, UK; E-Mails: (A.P.); (M.E.A.); (S.L.); (S.L.); (M.R.M.)
| | - Marcus R. Makowski
- Division of Imaging Sciences and Biomedical Engineering, King’s College London, 4th Floor, Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, UK; E-Mails: (A.P.); (M.E.A.); (S.L.); (S.L.); (M.R.M.)
- Department of Radiology, Charite, Berlin 10117, Germany
| | - René M. Botnar
- Division of Imaging Sciences and Biomedical Engineering, King’s College London, 4th Floor, Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, UK; E-Mails: (A.P.); (M.E.A.); (S.L.); (S.L.); (M.R.M.)
- Wellcome Trust and ESPRC Medical Engineering Center, King’s College London, London SE1 7EH, UK
- BHF Centre of Excellence, King’s College London, London SE1 7EH, UK
- NIHR Biomedical Research Centre, King’s College London, London SE1 7EH, UK
| |
Collapse
|
147
|
Kim B, Yang J, Hwang M, Choi J, Kim HO, Jang E, Lee JH, Ryu SH, Suh JS, Huh YM, Haam S. Aptamer-modified magnetic nanoprobe for molecular MR imaging of VEGFR2 on angiogenic vasculature. NANOSCALE RESEARCH LETTERS 2013; 8:399. [PMID: 24066922 PMCID: PMC3849016 DOI: 10.1186/1556-276x-8-399] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 09/09/2013] [Indexed: 05/17/2023]
Abstract
Nucleic acid-based aptamers have been developed for the specific delivery of diagnostic nanoprobes. Here, we introduce a new class of smart imaging nanoprobe, which is based on hybridization of a magnetic nanocrystal with a specific aptamer for specific detection of the angiogenic vasculature of glioblastoma via magnetic resonance (MR) imaging. The magnetic nanocrystal imaging core was synthesized using the thermal decomposition method and enveloped by carboxyl polysorbate 80 for water solubilization and conjugation of the targeting moiety. Subsequently, the surface of the carboxylated magnetic nanocrystal was modified with amine-functionalized aptamers that specifically bind to the vascular growth factor receptor 2 (VEGFR2) that is overexpressed on angiogenic vessels. To assess the targeted imaging potential of the aptamer-conjugated magnetic nanocrystal for VEGFR2 markers, the magnetic properties and MR imaging sensitivity were investigated using the orthotopic glioblastoma mouse model. In in vivo tests, the aptamer-conjugated magnetic nanocrystal effectively targeted VEGFR2 and demonstrated excellent MR imaging sensitivity with no cytotoxicity.
Collapse
Affiliation(s)
- Bongjune Kim
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, Seoul 120-749, Republic of Korea
| | - Jaemoon Yang
- Department of Radiology, College of Medicine, Yonsei University, Seoul 120-752, Republic of Korea
| | - Myeonghwan Hwang
- Department of Radiology, College of Medicine, Yonsei University, Seoul 120-752, Republic of Korea
| | - Jihye Choi
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, Seoul 120-749, Republic of Korea
| | - Hyun-Ouk Kim
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, Seoul 120-749, Republic of Korea
| | - Eunji Jang
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, Seoul 120-749, Republic of Korea
| | - Jung Hwan Lee
- POSTECH Aptamer Initiative Program, Division of Integrative Bioscience and Biotechnology, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Sung-Ho Ryu
- POSTECH Aptamer Initiative Program, Division of Integrative Bioscience and Biotechnology, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Jin-Suck Suh
- Department of Radiology, College of Medicine, Yonsei University, Seoul 120-752, Republic of Korea
| | - Yong-Min Huh
- Department of Radiology, College of Medicine, Yonsei University, Seoul 120-752, Republic of Korea
| | - Seungjoo Haam
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, Seoul 120-749, Republic of Korea
| |
Collapse
|
148
|
Phinikaridou A, Andia ME, Passacquale G, Ferro A, Botnar RM. Noninvasive MRI monitoring of the effect of interventions on endothelial permeability in murine atherosclerosis using an albumin-binding contrast agent. J Am Heart Assoc 2013; 2:e000402. [PMID: 24072533 PMCID: PMC3835253 DOI: 10.1161/jaha.113.000402] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Background Endothelial dysfunction promotes atherosclerosis. We investigated whether in vivo magnetic resonance imaging (MRI) using an albumin‐binding contrast agent, gadofosveset, could monitor the efficacy of minocycline and ebselen in reducing endothelial permeability and atherosclerotic burden in the brachiocephalic artery of high‐fat diet (HFD)–fed ApoE−/− mice. Methods and Results ApoE−/− mice were scanned 12 weeks after commencement of either a normal diet (controls) or an HFD. HFD‐fed ApoE−/− mice were either untreated or treated with minocycline or ebselen for 12 weeks. Delayed‐enhancement MRI and T1 mapping of the brachiocephalic artery, 30 minutes after injection of gadofosveset, showed increased vessel wall enhancement and relaxation rate (R1, s−1) in untreated HFD‐fed ApoE−/− mice (R1=3.8±0.52 s−1) compared with controls (R1=2.15±0.34 s−1, P<0.001). Conversely, minocycline‐treated (R1=2.7±0.17 s−1, P<0.001) and ebselen‐treated (R1=2.7±0.23 s−1, P<0.001) ApoE−/− mice showed less vessel wall enhancement compared with untreated HFD‐fed ApoE−/− mice. Mass spectroscopy showed a lower gadolinium concentration in the brachiocephalic artery of treated (minocycline=28.5±3 μmol/L, ebselen=32.4±4 μmol/L) compared with untreated HFD‐fed ApoE−/− mice (191±4.8 μmol/L) (P<0.02). Both interventions resulted in a lower plaque burden as measured by delayed‐enhancement MRI (minocycline=0.14±0.02 mm2, ebselen=0.20±0.09 mm2, untreated=0.44±0.01 mm2; P<0.001) and histology (minocycline=0.13±0.05 mm2, ebselen=0.18±0.02 mm2, untreated=0.32±0.04 mm2; P<0.002). Endothelium cells displayed fewer structural changes and smaller gap junction width in treated compared with untreated animals as seen by electron microscopy (minocycline=42.3±8.4 nm, ebselen=56.5±17 nm, untreated=2400±39 nm; P<0.001). Tissue flow cytometry of the brachiocephalic artery showed lower monocyte/macrophage content in both ebselen‐ and minocycline‐treated mice (8.06±3.2% and 7.62±1.73%, respectively) compared with untreated animals (20.1±2.2%) (P=0.03), with significant attenuation of the proinflammatory Ly6Chigh subtype (untreated mice, 42.64±6.1% of total monocytes; ebselen, 14.07±9.5% of total monocytes; minocycline, 26.42±0.6% of total monocytes). Conclusions We demonstrate that contrast‐enhanced MRI with an albumin‐binding contrast agent can be used to noninvasively monitor the effect of interventions on endothelial permeability and plaque burden. Blood albumin leakage could be a surrogate marker for the in vivo evaluation of interventions that aim at restoring endothelial integrity.
Collapse
Affiliation(s)
- Alkystis Phinikaridou
- Division of Imaging Science and Biomedical Engineering, King's College London, London, United Kingdom
| | | | | | | | | |
Collapse
|
149
|
Kitagawa T, Kosuge H, Chang E, James ML, Yamamoto T, Shen B, Chin FT, Gambhir SS, Dalman RL, McConnell MV. Integrin-targeted molecular imaging of experimental abdominal aortic aneurysms by (18)F-labeled Arg-Gly-Asp positron-emission tomography. Circ Cardiovasc Imaging 2013; 6:950-6. [PMID: 23995363 DOI: 10.1161/circimaging.113.000234] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Both inflammation and neoangiogenesis contribute to abdominal aortic aneurysm (AAA) disease. Arg-Gly-Asp-based molecular imaging has been shown to detect the integrin αvβ3. We studied a clinical dimeric (18)F-labeled Arg-Gly-Asp positron-emission tomography (PET) agent ((18)F-FPPRGD2) for molecular imaging of experimental AAAs. METHODS AND RESULTS Murine AAAs were induced in Apo-E-deficient mice by angiotensin II infusion, with monitoring of aortic diameter on ultrasound. AAA (n=10) and saline-infused control mice (n=7) were injected intravenously with (18)F-FPPRGD2, as well as an intravascular computed tomography contrast agent, then scanned using a small-animal PET/computed tomography scanner. Aortic uptake of (18)F-FPPRGD2 was quantified by percentage-injected dose per gram and target-to- BACKGROUND =0.003; median target-to- BACKGROUND =0.0008). Ex vivo autoradiography demonstrated high uptake of (18)F-FPPRGD2 into the AAA wall, with immunohistochemistry showing substantial cluster of differentiation (CD)-11b(+) macrophages and CD-31(+) neovessels. Target-to- BACKGROUND =-0.29, P=0.41) but did strongly correlate with both mural macrophage density (r=0.79, P=0.007) and neovessel counts (r=0.87, P=0.001) on immunohistochemistry. CONCLUSIONS PET imaging of experimental AAAs using (18)F-FPPRGD2 detects biologically active disease, correlating to the degree of vascular inflammation and neoangiogenesis. This may provide a clinically translatable molecular imaging approach to characterize AAA biology to predict risk beyond size alone.
Collapse
|
150
|
Quantitative evaluation using the plaque/muscle ratio index panels predicts plaque type and risk of embolism in patients undergoing carotid artery stenting. Clin Neurol Neurosurg 2013; 115:1298-303. [DOI: 10.1016/j.clineuro.2012.12.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Revised: 11/05/2012] [Accepted: 12/02/2012] [Indexed: 11/23/2022]
|