Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
JACC Case Rep ; 4(12): 704-705, 2022 Jun 15.
Article in English | MEDLINE | ID: mdl-35734524

ABSTRACT

Late complications in surgically corrected ALCAPA patients are rare. We describe an interesting case of a patient with a thrombosed giant right coronary artery aneurysm which was discovered on a chest X-ray. (Level of Difficulty: Intermediate.).

2.
Ann Surg ; 272(6): 919-924, 2020 12.
Article in English | MEDLINE | ID: mdl-33021367

ABSTRACT

OBJECTIVE: To determine the yield of preoperative screening for COVID-19 with chest CT and RT-PCR in patients without COVID-19 symptoms. SUMMARY OF BACKGROUND DATA: Many centers are currently screening surgical patients for COVID-19 using either chest CT, RT-PCR or both, due to the risk for worsened surgical outcomes and nosocomial spread. The optimal design and yield of such a strategy are currently unknown. METHODS: This multicenter study included consecutive adult patients without COVID-19 symptoms who underwent preoperative screening using chest CT and RT-PCR before elective or emergency surgery under general anesthesia. RESULTS: A total of 2093 patients without COVID-19 symptoms were included in 14 participating centers; 1224 were screened by CT and RT-PCR and 869 by chest CT only. The positive yield of screening using a combination of chest CT and RT-PCR was 1.5% [95% confidence interval (CI): 0.8-2.1]. Individual yields were 0.7% (95% CI: 0.2-1.1) for chest CT and 1.1% (95% CI: 0.6-1.7) for RT-PCR; the incremental yield of chest CT was 0.4%. In relation to COVID-19 community prevalence, up to ∼6% positive RT-PCR was found for a daily hospital admission rate >1.5 per 100,000 inhabitants, and around 1.0% for lower prevalence. CONCLUSIONS: One in every 100 patients without COVID-19 symptoms tested positive for SARS-CoV-2 with RT-PCR; this yield increased in conjunction with community prevalence. The added value of chest CT was limited. Preoperative screening allowed us to take adequate precautions for SARS-CoV-2 positive patients in a surgical population, whereas negative patients needed only routine procedures.


Subject(s)
Asymptomatic Infections , COVID-19/diagnosis , Emergency Treatment , Mass Screening/statistics & numerical data , Preoperative Care/methods , Reverse Transcriptase Polymerase Chain Reaction , SARS-CoV-2 , Surgical Procedures, Operative , Thorax/diagnostic imaging , Tomography, X-Ray Computed , Elective Surgical Procedures , Humans , Retrospective Studies
3.
Bioconjug Chem ; 31(2): 360-368, 2020 02 19.
Article in English | MEDLINE | ID: mdl-31095372

ABSTRACT

Nanotherapy has recently emerged as an experimental treatment option for atherosclerosis. To fulfill its promise, robust noninvasive imaging approaches for subject selection and treatment evaluation are warranted. To that end, we present here a positron emission tomography (PET)-based method for quantification of liposomal nanoparticle uptake in the atherosclerotic vessel wall. We evaluated a modular procedure to label liposomal nanoparticles with the radioisotope zirconium-89 (89Zr). Their biodistribution and vessel wall targeting in a rabbit atherosclerosis model was evaluated up to 15 days after intravenous injection by PET/computed tomography (CT) and PET/magnetic resonance imaging (PET/MRI). Vascular permeability was assessed in vivo using three-dimensional dynamic contrast-enhanced MRI (3D DCE-MRI) and ex vivo using near-infrared fluorescence (NIRF) imaging. The 89Zr-radiolabeled liposomes displayed a biodistribution pattern typical of long-circulating nanoparticles. Importantly, they markedly accumulated in atherosclerotic lesions in the abdominal aorta, as evident on PET/MRI and confirmed by autoradiography, and this uptake moderately correlated with vascular permeability. The method presented herein facilitates the development of nanotherapy for atherosclerotic disease as it provides a tool to screen for nanoparticle targeting in individual subjects' plaques.


Subject(s)
Atherosclerosis/diagnostic imaging , Liposomes/analysis , Plaque, Atherosclerotic/diagnostic imaging , Positron-Emission Tomography/methods , Radioisotopes/analysis , Zirconium/analysis , Animals , Aorta, Abdominal/diagnostic imaging , Male , Rabbits , Tissue Distribution
4.
Sci Transl Med ; 11(506)2019 08 21.
Article in English | MEDLINE | ID: mdl-31434756

ABSTRACT

Nanomedicine research produces hundreds of studies every year, yet very few formulations have been approved for clinical use. This is due in part to a reliance on murine studies, which have limited value in accurately predicting translational efficacy in larger animal models and humans. Here, we report the scale-up of a nanoimmunotherapy from mouse to large rabbit and porcine atherosclerosis models, with an emphasis on the solutions we implemented to overcome production and evaluation challenges. Specifically, we integrated translational imaging readouts within our workflow to both analyze the nanoimmunotherapeutic's in vivo behavior and assess treatment response in larger animals. We observed our nanoimmunotherapeutic's anti-inflammatory efficacy in mice, as well as rabbits and pigs. Nanoimmunotherapy-mediated reduction of inflammation in the large animal models halted plaque progression, supporting the approach's translatability and potential to acutely treat atherosclerosis.


Subject(s)
Atherosclerosis/immunology , Atherosclerosis/therapy , Imaging, Three-Dimensional , Immunotherapy , Nanomedicine , Animals , Apolipoproteins E/deficiency , Atherosclerosis/diagnostic imaging , Atherosclerosis/drug therapy , Disease Models, Animal , Female , Lipoproteins, HDL/metabolism , Lipoproteins, HDL/toxicity , Magnetic Resonance Imaging , Male , Mice, Inbred C57BL , Positron-Emission Tomography , Rabbits , Simvastatin/pharmacology , Simvastatin/therapeutic use , Species Specificity , Swine , Tissue Distribution
5.
JACC Cardiovasc Imaging ; 12(10): 2015-2026, 2019 10.
Article in English | MEDLINE | ID: mdl-30343086

ABSTRACT

OBJECTIVES: This study sought to develop an integrative positron emission tomography (PET) with magnetic resonance imaging (MRI) procedure for accurate atherosclerotic plaque phenotyping, facilitated by clinically approved and nanobody radiotracers. BACKGROUND: Noninvasive characterization of atherosclerosis remains a challenge in clinical practice. The limitations of current diagnostic methods demonstrate that, in addition to atherosclerotic plaque morphology and composition, disease activity needs to be evaluated. METHODS: We screened 3 nanobody radiotracers targeted to different biomarkers of atherosclerosis progression, namely vascular cell adhesion molecule (VCAM)-1, lectin-like oxidized low-density lipoprotein receptor (LOX)-1, and macrophage mannose receptor (MMR). The nanobodies, initially radiolabeled with copper-64 (64Cu), were extensively evaluated in Apoe-/- mice and atherosclerotic rabbits using a combination of in vivo PET/MRI readouts and ex vivo radioactivity counting, autoradiography, and histological analyses. RESULTS: The 3 nanobody radiotracers accumulated in atherosclerotic plaques and displayed short circulation times due to fast renal clearance. The MMR nanobody was selected for labeling with gallium-68 (68Ga), a short-lived radioisotope with high clinical relevance, and used in an ensuing atherosclerosis progression PET/MRI study. Macrophage burden was longitudinally studied by 68Ga-MMR-PET, plaque burden by T2-weighted MRI, and neovascularization by dynamic contrast-enhanced (DCE) MRI. Additionally, inflammation and microcalcifications were evaluated by fluorine-18 (18F)-labeled fluorodeoxyglucose (18F-FDG) and 18F-sodium fluoride (18F-NaF) PET, respectively. We observed an increase in all the aforementioned measures as disease progressed, and the imaging signatures correlated with histopathological features. CONCLUSIONS: We have evaluated nanobody-based radiotracers in rabbits and developed an integrative PET/MRI protocol that allows noninvasive assessment of different processes relevant to atherosclerosis progression. This approach allows the multiparametric study of atherosclerosis and can aid in early stage anti-atherosclerosis drug trials.


Subject(s)
Atherosclerosis/diagnostic imaging , Multiparametric Magnetic Resonance Imaging , Plaque, Atherosclerotic , Positron-Emission Tomography , Radiopharmaceuticals/administration & dosage , Single-Domain Antibodies/administration & dosage , Animals , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Disease Models, Animal , Disease Progression , Early Diagnosis , Genetic Predisposition to Disease , Lectins, C-Type/immunology , Mannose Receptor , Mannose-Binding Lectins/immunology , Mice, Knockout, ApoE , Multimodal Imaging , Phenotype , Rabbits , Radiopharmaceuticals/pharmacokinetics , Receptors, Cell Surface/immunology , Scavenger Receptors, Class E/immunology , Single-Domain Antibodies/metabolism , Vascular Cell Adhesion Molecule-1/immunology
6.
Methods Mol Biol ; 1816: 385-400, 2018.
Article in English | MEDLINE | ID: mdl-29987836

ABSTRACT

Several animal models have been developed to study atherosclerosis. Here we present a rabbit atherosclerosis model generated by surgical denudation of the aortic endothelium in combination with a high-fat and cholesterol-enriched diet. This model is characterized by the formation of vascular lesions that exhibit several hallmarks of human atherosclerosis. Due to the rabbit's relative large size, as compared to rodents, this model is suited for the imaging-guided evaluation of novel therapeutic strategies using clinical scanners. In this chapter, we present an extensive outline of the procedures to induce aortic atherosclerotic lesions in rabbits as well as methods to evaluate the disease, including noninvasive in vivo multiparametric imaging and histopathology.


Subject(s)
Atherosclerosis/etiology , Atherosclerosis/pathology , Disease Models, Animal , Rabbits , Animals , Aorta/diagnostic imaging , Aorta/pathology , Atherosclerosis/diagnostic imaging , Diet, High-Fat/adverse effects , Humans , Magnetic Resonance Imaging/methods , Male , Plaque, Atherosclerotic/diagnostic imaging , Plaque, Atherosclerotic/etiology , Plaque, Atherosclerotic/pathology , Positron-Emission Tomography/methods , Rabbits/physiology
7.
J Am Coll Cardiol ; 71(3): 321-335, 2018 01 23.
Article in English | MEDLINE | ID: mdl-29348025

ABSTRACT

BACKGROUND: Oxidation-specific epitopes (OSEs) are proinflammatory, and elevated levels in plasma predict cardiovascular events. OBJECTIVES: The purpose of this study was to develop novel positron emission tomography (PET) probes to noninvasively image OSE-rich lesions. METHODS: An antigen-binding fragment (Fab) antibody library was constructed from human fetal cord blood. After multiple rounds of screening against malondialdehyde-acetaldehyde (MAA) epitopes, the Fab LA25 containing minimal nontemplated insertions in the CDR3 region was identified and characterized. In mice, pharmacokinetics, biodistribution, and plaque specificity studies were performed with Zirconium-89 (89Zr)-labeled LA25. In rabbits, 89Zr-LA25 was used in combination with an integrated clinical PET/magnetic resonance (MR) system. 18F-fluorodeoxyglucose PET and dynamic contrast-enhanced MR imaging were used to evaluate vessel wall inflammation and plaque neovascularization, respectively. Extensive ex vivo validation was carried out through a combination of gamma counting, near infrared fluorescence, autoradiography, immunohistochemistry, and immunofluorescence. RESULTS: LA25 bound specifically to MAA epitopes in advanced and ruptured human atherosclerotic plaques with accompanying thrombi and in debris from distal protection devices. PET/MR imaging 24 h after injection of 89Zr-LA25 showed increased uptake in the abdominal aorta of atherosclerotic rabbits compared with nonatherosclerotic control rabbits, confirmed by ex vivo gamma counting and autoradiography. 18F-fluorodeoxyglucose PET, dynamic contrast-enhanced MR imaging, and near-infrared fluorescence signals were also significantly higher in atherosclerotic rabbit aortas compared with control aortas. Enhanced liver uptake was also noted in atherosclerotic animals, confirmed by the presence of MAA epitopes by immunostaining. CONCLUSIONS: 89Zr-LA25 is a novel PET radiotracer that may allow noninvasive phenotyping of high-risk OSE-rich lesions.


Subject(s)
Acetaldehyde , Atherosclerosis/diagnostic imaging , Magnetic Resonance Imaging/methods , Malondialdehyde , Positron-Emission Tomography/methods , Thrombosis/diagnostic imaging , Acetaldehyde/metabolism , Animals , Atherosclerosis/metabolism , Epitopes/metabolism , Humans , Malondialdehyde/metabolism , Mice , Mice, Knockout , Rabbits , Thrombosis/metabolism , Tissue Distribution/physiology
8.
JACC Cardiovasc Imaging ; 9(8): 950-61, 2016 08.
Article in English | MEDLINE | ID: mdl-27236528

ABSTRACT

OBJECTIVES: The goal of this study was to develop and validate a noninvasive imaging tool to visualize the in vivo behavior of high-density lipoprotein (HDL) by using positron emission tomography (PET), with an emphasis on its plaque-targeting abilities. BACKGROUND: HDL is a natural nanoparticle that interacts with atherosclerotic plaque macrophages to facilitate reverse cholesterol transport. HDL-cholesterol concentration in blood is inversely associated with risk of coronary heart disease and remains one of the strongest independent predictors of incident cardiovascular events. METHODS: Discoidal HDL nanoparticles were prepared by reconstitution of its components apolipoprotein A-I (apo A-I) and the phospholipid 1,2-dimyristoyl-sn-glycero-3-phosphocholine. For radiolabeling with zirconium-89 ((89)Zr), the chelator deferoxamine B was introduced by conjugation to apo A-I or as a phospholipid-chelator (1,2-distearoyl-sn-glycero-3-phosphoethanolamine-deferoxamine B). Biodistribution and plaque targeting of radiolabeled HDL were studied in established murine, rabbit, and porcine atherosclerosis models by using PET combined with computed tomography (PET/CT) imaging or PET combined with magnetic resonance imaging. Ex vivo validation was conducted by radioactivity counting, autoradiography, and near-infrared fluorescence imaging. Flow cytometric assessment of cellular specificity in different tissues was performed in the murine model. RESULTS: We observed distinct pharmacokinetic profiles for the two (89)Zr-HDL nanoparticles. Both apo A-I- and phospholipid-labeled HDL mainly accumulated in the kidneys, liver, and spleen, with some marked quantitative differences in radioactivity uptake values. Radioactivity concentrations in rabbit atherosclerotic aortas were 3- to 4-fold higher than in control animals at 5 days' post-injection for both (89)Zr-HDL nanoparticles. In the porcine model, increased accumulation of radioactivity was observed in lesions by using in vivo PET imaging. Irrespective of the radiolabel's location, HDL nanoparticles were able to preferentially target plaque macrophages and monocytes. CONCLUSIONS: (89)Zr labeling of HDL allows study of its in vivo behavior by using noninvasive PET imaging, including visualization of its accumulation in advanced atherosclerotic lesions. The different labeling strategies provide insight on the pharmacokinetics and biodistribution of HDL's main components (i.e., phospholipids, apo A-I).


Subject(s)
Aorta/diagnostic imaging , Aortic Diseases/diagnostic imaging , Atherosclerosis/diagnostic imaging , Lipoproteins, HDL/administration & dosage , Magnetic Resonance Imaging , Molecular Imaging/methods , Plaque, Atherosclerotic , Positron Emission Tomography Computed Tomography , Radiopharmaceuticals/administration & dosage , Zirconium/administration & dosage , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Autoradiography , Disease Models, Animal , Female , Flow Cytometry , Lipoproteins, HDL/pharmacokinetics , Male , Mice, Inbred C57BL , Mice, Knockout , Optical Imaging , Predictive Value of Tests , Rabbits , Radioisotopes , Radiopharmaceuticals/pharmacokinetics , Reproducibility of Results , Tissue Distribution , Zirconium/pharmacokinetics
9.
NMR Biomed ; 28(10): 1304-14, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26332103

ABSTRACT

Atherosclerotic plaques that cause stroke and myocardial infarction are characterized by increased microvascular permeability and inflammation. Dynamic contrast-enhanced MRI (DCE-MRI) has been proposed as a method to quantify vessel wall microvascular permeability in vivo. Until now, most DCE-MRI studies of atherosclerosis have been limited to two-dimensional (2D) multi-slice imaging. Although providing the high spatial resolution required to image the arterial vessel wall, these approaches do not allow the quantification of plaque permeability with extensive anatomical coverage, an essential feature when imaging heterogeneous diseases, such as atherosclerosis. To our knowledge, we present the first systematic evaluation of three-dimensional (3D), high-resolution, DCE-MRI for the extensive quantification of plaque permeability along an entire vascular bed, with validation in atherosclerotic rabbits. We compare two acquisitions: 3D turbo field echo (TFE) with motion-sensitized-driven equilibrium (MSDE) preparation and 3D turbo spin echo (TSE). We find 3D TFE DCE-MRI to be superior to 3D TSE DCE-MRI in terms of temporal stability metrics. Both sequences show good intra- and inter-observer reliability, and significant correlation with ex vivo permeability measurements by Evans Blue near-infrared fluorescence (NIRF). In addition, we explore the feasibility of using compressed sensing to accelerate 3D DCE-MRI of atherosclerosis, to improve its temporal resolution and therefore the accuracy of permeability quantification. Using retrospective under-sampling and reconstructions, we show that compressed sensing alone may allow the acceleration of 3D DCE-MRI by up to four-fold. We anticipate that the development of high-spatial-resolution 3D DCE-MRI with prospective compressed sensing acceleration may allow for the more accurate and extensive quantification of atherosclerotic plaque permeability along an entire vascular bed. We foresee that this approach may allow for the comprehensive and accurate evaluation of plaque permeability in patients, and may be a useful tool to assess the therapeutic response to approved and novel drugs for cardiovascular disease.


Subject(s)
Aorta, Abdominal/pathology , Aortic Diseases/pathology , Capillary Permeability , Image Processing, Computer-Assisted/methods , Imaging, Three-Dimensional/methods , Magnetic Resonance Imaging/methods , Plaque, Atherosclerotic/pathology , Animals , Contrast Media , Disease Models, Animal , Feasibility Studies , Observer Variation , Rabbits
10.
Sci Adv ; 1(3)2015 Apr.
Article in English | MEDLINE | ID: mdl-26295063

ABSTRACT

Inflammation drives atherosclerotic plaque progression and rupture, and is a compelling therapeutic target. Consequently, attenuating inflammation by reducing local macrophage accumulation is an appealing approach. This can potentially be accomplished by either blocking blood monocyte recruitment to the plaque or increasing macrophage apoptosis and emigration. Because macrophage proliferation was recently shown to dominate macrophage accumulation in advanced plaques, locally inhibiting macrophage proliferation may reduce plaque inflammation and produce long-term therapeutic benefits. To test this hypothesis, we used nanoparticle-based delivery of simvastatin to inhibit plaque macrophage proliferation in apolipoprotein E deficient mice (Apoe-/- ) with advanced atherosclerotic plaques. This resulted in rapid reduction of plaque inflammation and favorable phenotype remodeling. We then combined this short-term nanoparticle intervention with an eight-week oral statin treatment, and this regimen rapidly reduced and continuously suppressed plaque inflammation. Our results demonstrate that pharmacologically inhibiting local macrophage proliferation can effectively treat inflammation in atherosclerosis.

11.
Nanomedicine ; 11(5): 1133-40, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25791805

ABSTRACT

The present study describes the development of a good manufacturing practice (GMP)-grade liposomal nanotherapy containing prednisolone phosphate for the treatment of inflammatory diseases. After formulation design, GMP production was commenced which yielded consistent, stable liposomes sized 100nm±10nm, with a prednisolone phosphate (PLP) incorporation efficiency of 3%-5%. Pharmacokinetics and toxicokinetics of GMP-grade liposomal nanoparticles were evaluated in healthy rats, which were compared to daily and weekly administration of free prednisolone phosphate, revealing a long circulatory half-life with minimal side effects. Subsequently, non-invasive multimodal clinical imaging after liposomal nanotherapy's intravenous administration revealed anti-inflammatory effects on the vessel wall of atherosclerotic rabbits. The present program led to institutional review board approval for two clinical trials with patients with atherosclerosis. FROM THE CLINICAL EDITOR: In drug discovery, bringing production to industrial scale is an essential process. In this article the authors describe the development of an anti-inflammatory nanoparticle according to good manufacturing practice. As a result, this paves the way for translating laboratory studies to clinical trials in humans.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Atherosclerosis/drug therapy , Chemistry, Pharmaceutical/methods , Glucocorticoids/administration & dosage , Prednisolone/analogs & derivatives , Animals , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/toxicity , Aorta/drug effects , Aorta/pathology , Atherosclerosis/pathology , Glucocorticoids/pharmacokinetics , Glucocorticoids/therapeutic use , Glucocorticoids/toxicity , Half-Life , Humans , Liposomes , Male , Prednisolone/administration & dosage , Prednisolone/pharmacokinetics , Prednisolone/therapeutic use , Prednisolone/toxicity , Rabbits , Rats , Rats, Sprague-Dawley , Rats, Wistar
12.
Nanomedicine ; 11(5): 1039-46, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25791806

ABSTRACT

Drug delivery to atherosclerotic plaques via liposomal nanoparticles may improve therapeutic agents' risk-benefit ratios. Our paper details the first clinical studies of a liposomal nanoparticle encapsulating prednisolone (LN-PLP) in atherosclerosis. First, PLP's liposomal encapsulation improved its pharmacokinetic profile in humans (n=13) as attested by an increased plasma half-life of 63h (LN-PLP 1.5mg/kg). Second, intravenously infused LN-PLP appeared in 75% of the macrophages isolated from iliofemoral plaques of patients (n=14) referred for vascular surgery in a randomized, placebo-controlled trial. LN-PLP treatment did however not reduce arterial wall permeability or inflammation in patients with atherosclerotic disease (n=30), as assessed by multimodal imaging in a subsequent randomized, placebo-controlled study. In conclusion, we successfully delivered a long-circulating nanoparticle to atherosclerotic plaque macrophages in patients, whereas prednisolone accumulation in atherosclerotic lesions had no anti-inflammatory effect. Nonetheless, the present study provides guidance for development and imaging-assisted evaluation of future nanomedicine in atherosclerosis. FROM THE CLINICAL EDITOR: In this study, the authors undertook the first clinical trial using long-circulating liposomal nanoparticle encapsulating prednisolone in patients with atherosclerosis, based on previous animal studies. Despite little evidence of anti-inflammatory effect, the results have provided a starting point for future development of nanomedicine in cardiovascular diseases.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Atherosclerosis/drug therapy , Glucocorticoids/administration & dosage , Macrophages/drug effects , Plaque, Atherosclerotic/drug therapy , Prednisolone/administration & dosage , Administration, Intravenous , Adult , Aged , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/therapeutic use , Arteries/drug effects , Arteries/pathology , Atherosclerosis/pathology , Female , Glucocorticoids/pharmacokinetics , Glucocorticoids/therapeutic use , Humans , Liposomes , Macrophages/pathology , Male , Middle Aged , Plaque, Atherosclerotic/pathology , Prednisolone/pharmacokinetics , Prednisolone/therapeutic use
13.
Bioconjug Chem ; 26(3): 443-51, 2015 Mar 18.
Article in English | MEDLINE | ID: mdl-25650634

ABSTRACT

High-density lipoprotein (HDL) is a natural nanoparticle that exhibits an intrinsic affinity for atherosclerotic plaque macrophages. Its natural targeting capability as well as the option to incorporate lipophilic payloads, e.g., imaging or therapeutic components, in both the hydrophobic core and the phospholipid corona make the HDL platform an attractive nanocarrier. To realize controlled release properties, we developed a hybrid polymer/HDL nanoparticle composed of a lipid/apolipoprotein coating that encapsulates a poly(lactic-co-glycolic acid) (PLGA) core. This novel HDL-like nanoparticle (PLGA-HDL) displayed natural HDL characteristics, including preferential uptake by macrophages and a good cholesterol efflux capacity, combined with a typical PLGA nanoparticle slow release profile. In vivo studies carried out with an ApoE knockout mouse model of atherosclerosis showed clear accumulation of PLGA-HDL nanoparticles in atherosclerotic plaques, which colocalized with plaque macrophages. This biomimetic platform integrates the targeting capacity of HDL biomimetic nanoparticles with the characteristic versatility of PLGA-based nanocarriers.


Subject(s)
Biomimetic Materials/metabolism , Lactic Acid/metabolism , Lipoproteins, HDL/metabolism , Macrophages/metabolism , Nanoparticles/metabolism , Plaque, Atherosclerotic/metabolism , Polyglycolic Acid/metabolism , Animals , Biomimetic Materials/administration & dosage , Biomimetic Materials/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Drug Delivery Systems/methods , Human Umbilical Vein Endothelial Cells , Humans , Lactic Acid/administration & dosage , Lactic Acid/chemistry , Lipoproteins, HDL/administration & dosage , Lipoproteins, HDL/chemistry , Macrophages/drug effects , Macrophages/pathology , Male , Mice , Mice, Knockout , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Plaque, Atherosclerotic/drug therapy , Plaque, Atherosclerotic/pathology , Polyglycolic Acid/administration & dosage , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer
14.
ACS Nano ; 9(2): 1837-47, 2015 Feb 24.
Article in English | MEDLINE | ID: mdl-25619964

ABSTRACT

Atherosclerosis is a major cause of global morbidity and mortality that could benefit from novel targeted therapeutics. Recent studies have shown efficient and local drug delivery with nanoparticles, although the nanoparticle targeting mechanism for atherosclerosis has not yet been fully elucidated. Here we used in vivo and ex vivo multimodal imaging to examine permeability of the vessel wall and atherosclerotic plaque accumulation of fluorescently labeled liposomal nanoparticles in a rabbit model. We found a strong correlation between permeability as established by in vivo dynamic contrast enhanced magnetic resonance imaging and nanoparticle plaque accumulation with subsequent nanoparticle distribution throughout the vessel wall. These key observations will enable the development of nanotherapeutic strategies for atherosclerosis.


Subject(s)
Multimodal Imaging/methods , Nanoparticles , Plaque, Atherosclerotic/blood , Plaque, Atherosclerotic/diagnosis , Animals , Capillary Permeability , Fluorescent Dyes/chemistry , Male , Nanoparticles/chemistry , Nanoparticles/metabolism , Plaque, Atherosclerotic/metabolism , Rabbits , Time Factors
15.
J Am Coll Cardiol ; 64(14): 1418-26, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-25277610

ABSTRACT

BACKGROUND: Patients with familial hypercholesterolemia (FH) are characterized by elevated atherogenic lipoprotein particles, predominantly low-density lipoprotein cholesterol (LDL-C), which is associated with accelerated atherogenesis and increased cardiovascular risk. OBJECTIVES: This study used (18)F-fluorodeoxyglucose positron emission tomography ((18)FDG-PET) to investigate whether arterial inflammation is higher in patients with FH and, moreover, whether lipoprotein apheresis attenuates arterial wall inflammation in FH patients. METHODS: In total, 38 subjects were recruited: 24 FH patients and 14 normolipidemic controls. All subjects underwent FDG-PET imaging at baseline. Twelve FH patients who met the criteria for lipoprotein apheresis underwent apheresis procedures followed by a second FDG-PET imaging 3 days (range 1 to 4 days) after apheresis. Subsequently, the target-to-background ratio (TBR) of FDG uptake within the arterial wall was assessed. RESULTS: In FH patients, the mean arterial TBR was higher compared with healthy controls (2.12 ± 0.27 vs. 1.92 ± 0.19; p = 0.03). A significant correlation was observed between baseline arterial TBR and LDL-C (R = 0.37; p = 0.03) that remained significant after adjusting for statin use (ß = 0.001; p = 0.02) and atherosclerosis risk factors (ß = 0.001; p = 0.03). LDL-C levels were significantly reduced after lipoprotein apheresis (284 ± 118 mg/dl vs. 127 ± 50 mg/dl; p < 0.001). There was a significant reduction of arterial inflammation after lipoprotein apheresis (TBR: 2.05 ± 0.31 vs. 1.91 ± 0.33; p < 0.02). CONCLUSIONS: The arterial wall of FH patients is characterized by increased inflammation, which is markedly reduced after lipoprotein apheresis. This lends support to a causal role of apoprotein B-containing lipoproteins in arterial wall inflammation and supports the concept that lipoprotein-lowering therapies may impart anti-inflammatory effects by reducing atherogenic lipoproteins.


Subject(s)
Arteries/pathology , Blood Component Removal/methods , Hyperlipoproteinemia Type II/therapy , Inflammation/therapy , Aged , Atherosclerosis/blood , Case-Control Studies , Cholesterol, LDL/blood , Cross-Sectional Studies , Female , Fluorodeoxyglucose F18/chemistry , Humans , Hyperlipoproteinemia Type II/complications , Inflammation/complications , Lipoproteins/chemistry , Male , Middle Aged , Pilot Projects , Positron-Emission Tomography/methods , Prospective Studies , Risk Factors , Tomography, X-Ray Computed
16.
Nat Commun ; 5: 3065, 2014.
Article in English | MEDLINE | ID: mdl-24445279

ABSTRACT

Inflammation is a key feature of atherosclerosis and a target for therapy. Statins have potent anti-inflammatory properties but these cannot be fully exploited with oral statin therapy due to low systemic bioavailability. Here we present an injectable reconstituted high-density lipoprotein (rHDL) nanoparticle carrier vehicle that delivers statins to atherosclerotic plaques. We demonstrate the anti-inflammatory effect of statin-rHDL in vitro and show that this effect is mediated through the inhibition of the mevalonate pathway. We also apply statin-rHDL nanoparticles in vivo in an apolipoprotein E-knockout mouse model of atherosclerosis and show that they accumulate in atherosclerotic lesions in which they directly affect plaque macrophages. Finally, we demonstrate that a 3-month low-dose statin-rHDL treatment regimen inhibits plaque inflammation progression, while a 1-week high-dose regimen markedly decreases inflammation in advanced atherosclerotic plaques. Statin-rHDL represents a novel potent atherosclerosis nanotherapy that directly affects plaque inflammation.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Lipoproteins, HDL/therapeutic use , Nanoparticles/therapeutic use , Plaque, Atherosclerotic/prevention & control , Administration, Intravenous , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Disease Models, Animal , Dose-Response Relationship, Drug , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Lipoproteins, HDL/administration & dosage , Male , Mice , Mice, Knockout , Nanoparticles/administration & dosage , Plaque, Atherosclerotic/genetics , Treatment Outcome
17.
Proc Natl Acad Sci U S A ; 111(3): 1078-83, 2014 Jan 21.
Article in English | MEDLINE | ID: mdl-24395808

ABSTRACT

Therapeutic and diagnostic nanomaterials are being intensely studied for several diseases, including cancer and atherosclerosis. However, the exact mechanism by which nanomedicines accumulate at targeted sites remains a topic of investigation, especially in the context of atherosclerotic disease. Models to accurately predict transvascular permeation of nanomedicines are needed to aid in design optimization. Here we show that an endothelialized microchip with controllable permeability can be used to probe nanoparticle translocation across an endothelial cell layer. To validate our in vitro model, we studied nanoparticle translocation in an in vivo rabbit model of atherosclerosis using a variety of preclinical and clinical imaging methods. Our results reveal that the translocation of lipid-polymer hybrid nanoparticles across the atherosclerotic endothelium is dependent on microvascular permeability. These results were mimicked with our microfluidic chip, demonstrating the potential utility of the model system.


Subject(s)
Atherosclerosis/physiopathology , Drug Delivery Systems , Endothelium/metabolism , Metal Nanoparticles/chemistry , Animals , Atherosclerosis/drug therapy , Disease Models, Animal , Gold/chemistry , Human Umbilical Vein Endothelial Cells , Humans , Magnetic Resonance Imaging , Male , Microcirculation , Microfluidics , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Models, Theoretical , Permeability , Plaque, Atherosclerotic , Rabbits , Shear Strength
18.
Methods Enzymol ; 508: 211-28, 2012.
Article in English | MEDLINE | ID: mdl-22449928

ABSTRACT

Nanomedicine can provide a potent alternative to current therapeutic strategies for atherosclerosis. For example, the encapsulation of anti-inflammatory drugs into liposomes improves their pharmacokinetics and biodistribution, thereby enhancing bioavailability to atherosclerotic plaques and improving therapeutic efficacy. The evaluation of this type of experimental therapeutics can greatly benefit from in vivo evaluation to assess biological changes, which can be performed by non-invasive imaging techniques, such as ¹8F-fluorodeoxyglucose positron emission tomography/computed tomography (FDG-PET/CT) and dynamic contrast enhanced magnetic resonance imaging (DCE-MRI). Here, we will illustrate the methods for inducing atherosclerosis in a rabbit model, the production of anti-inflammatory liposomes and monitoring of therapeutic efficacy of experimental therapeutics with the above-mentioned imaging techniques.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Atherosclerosis/drug therapy , Disease Models, Animal , Liposomes , Angioplasty, Balloon , Animals , Atherosclerosis/pathology , Magnetic Resonance Imaging , Multimodal Imaging , Positron-Emission Tomography , Rabbits , Tomography, X-Ray Computed
19.
Curr Cardiovasc Imaging Rep ; 5(1): 19-25, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22308199

ABSTRACT

Cardiovascular disease (CVD) is the leading cause of morbidity and mortality worldwide. New diagnostic and therapeutic strategies are needed to mitigate this public health issue. Advances in nanotechnology have generated innovative strategies for diagnosis and therapy in a variety of diseases, foremost in cancer. Based on these studies, a novel concept referred to as nanomedical theranostics, or the combinatory application of nanoparticulate agents to allow diagnostic therapy, is being explored to enable image-guided, personalized, or targeted treatment. Preclinically, theranostics have been gradually applied to CVD with several interesting and encouraging findings. This article summarizes studies and challenges of nanotheranostic strategies in CVD. It also evaluates nanotheranostic strategies that may potentially be utilized to benefit patients.

20.
J Am Coll Cardiol ; 58(24): 2522-30, 2011 Dec 06.
Article in English | MEDLINE | ID: mdl-22133853

ABSTRACT

OBJECTIVES: The goal of this study was to use noninvasive conventional and molecular magnetic resonance imaging (MRI) to detect and characterize abdominal aortic aneurysms (AAAs) in vivo. BACKGROUND: Collagen is an essential constituent of aneurysms. Noninvasive MRI of collagen may represent an opportunity to help detect and better characterize AAAs and initiate intervention. METHODS: We used an AAA C57BL/6 mouse model in which a combination of angiotensin II infusion and transforming growth factor-ß neutralization results in AAA formation with incidence of aortic rupture. High-resolution, multisequence MRI was performed to characterize the temporal progression of an AAA. To allow molecular MRI of collagen, paramagnetic/fluorescent micellar nanoparticles functionalized with a collagen-binding protein (CNA-35) were intravenously administered. In vivo imaging results were corroborated with immunohistochemistry and confocal fluorescence microscopy. RESULTS: High-resolution, multisequence MRI allowed the visualization of the primary fibrotic response in the aortic wall. As the aneurysm progressed, the formation of a secondary channel or dissection was detected. Further analysis revealed a dramatic increase of the aortic diameter. Injection of CNA-35 micelles resulted in a significantly higher magnetic resonance signal enhancement in the aneurysmal wall compared with nonspecific micelles. Histological studies revealed the presence of collagen in regions of magnetic resonance signal enhancement, and confocal microscopy proved the precise co-localization of CNA-35 micelles with type I collagen. In addition, in a proof-of-concept experiment, we reported the potential of CNA-35 micelles to discriminate between stable AAA lesions and aneurysms that were likely to rapidly progress or rupture. CONCLUSIONS: High-resolution, multisequence MRI allowed longitudinal monitoring of AAA progression while the presence of collagen was visualized by nanoparticle-enhanced MRI.


Subject(s)
Aortic Aneurysm, Abdominal/diagnosis , Disease Models, Animal , Magnetic Resonance Imaging , Animals , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Collagen/analysis , Contrast Media , Disease Progression , Image Enhancement , Immunohistochemistry , Magnetic Resonance Imaging/methods , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Transforming Growth Factor beta/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...