Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Claudia Calcagno is active.

Publication


Featured researches published by Claudia Calcagno.


Nano Letters | 2008

Nanocrystal core high-density lipoproteins: a multimodality contrast agent platform

David P. Cormode; Torjus Skajaa; Matti M. van Schooneveld; Rolf Koole; Peter A. Jarzyna; Mark E. Lobatto; Claudia Calcagno; Alessandra Barazza; Ronald E. Gordon; Pat Zanzonico; Edward A. Fisher; Zahi A. Fayad; Willem J. M. Mulder

High density lipoprotein (HDL) is an important natural nanoparticle that may be modified for biomedical imaging purposes. Here we developed a novel technique to create unique multimodality HDL mimicking nanoparticles by incorporation of gold, iron oxide, or quantum dot nanocrystals for computed tomography, magnetic resonance, and fluorescence imaging, respectively. By including additional labels in the corona of the particles, they were made multifunctional. The characteristics of these nanoparticles, as well as their in vitro and in vivo behavior, revealed that they closely mimic native HDL.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2008

Detection of Neovessels in Atherosclerotic Plaques of Rabbits Using Dynamic Contrast Enhanced MRI and 18F-FDG PET

Claudia Calcagno; Jean-Christophe Cornily; Fabien Hyafil; James H.F. Rudd; Karen C. Briley-Saebo; Venkatesh Mani; Gregg Goldschlager; Josef Machac; Valentin Fuster; Zahi A. Fayad

Objective—The association of inflammatory cells and neovessels in atherosclerosis is considered a histological hallmark of high-risk active lesions. Therefore, the development and validation of noninvasive imaging techniques that allow for the detection of inflammation and neoangiogenesis in atherosclerosis would be of major clinical interest. Our aim was to test 2 techniques, black blood dynamic contrast enhanced MRI (DCE-MRI) and 18-fluorine-fluorodeoxyglucose (18F-FDG) PET, to quantify inflammation expressed as plaque neovessels content in a rabbit model of atherosclerosis. Methods and Results—Atherosclerotic plaques were induced in the aorta of 10 rabbits by a combination of 2 endothelial abrasions and 4 months hyperlipidemic diet. Six rabbits underwent MRI during the injection of Gd-DTPA, whereas 4 rabbits were imaged after injection of 18F-FDG with PET. We found a positive correlation between neovessels count in atherosclerotic plaques and (1) Gd-DTPA uptake parameters evaluated by DCE-MRI (r=0.89, P=0.016) and (2) 18F-FDG uptake evaluated by PET (r=0.5, P=0.103 after clustered robust, Huber-White, standard errors analysis). Conclusion—DCE-MRI and 18F-FDG PET may allow for the evaluation of inflammation in atherosclerotic plaques of rabbits. These noninvasive imaging modalities could be proposed as clinical tools in the evaluation of lesion prognosis and monitoring of anti–angiogenic therapies.


Molecular Pharmaceutics | 2010

Multimodal clinical imaging to longitudinally assess a nanomedical anti-inflammatory treatment in experimental atherosclerosis.

Mark E. Lobatto; Zahi A. Fayad; Stephane Silvera; Esad Vucic; Claudia Calcagno; Venkatesh Mani; Stephen D. Dickson; Klaas Nicolay; Manuela Banciu; Raymond M. Schiffelers; Josbert M. Metselaar; Louis van Bloois; Hai-Shan Wu; John T. Fallon; James H.F. Rudd; Valentin Fuster; Edward A. Fisher; Gert Storm; Willem J. M. Mulder

Atherosclerosis is an inflammatory disease causing great morbidity and mortality in the Western world. To increase the anti-inflammatory action and decrease adverse effects of glucocorticoids (PLP), a nanomedicinal liposomal formulation of this drug (L-PLP) was developed and intravenously applied at a dose of 15 mg/kg PLP to a rabbit model of atherosclerosis. Since atherosclerosis is a systemic disease, emerging imaging modalities for assessing atherosclerotic plaque are being developed. (18)F-Fluoro-deoxy-glucose positron emission tomography and dynamic contrast enhanced magnetic resonance imaging, methods commonly used in oncology, were applied to longitudinally assess therapeutic efficacy. Significant anti-inflammatory effects were observed as early as 2 days that lasted up to at least 7 days after administration of a single dose of L-PLP. No significant changes were found for the free PLP treated animals. These findings were corroborated by immunohistochemical analysis of macrophage density in the vessel wall. In conclusion, this study evaluates a powerful two-pronged strategy for efficient treatment of atherosclerosis that includes nanomedical therapy of atherosclerotic plaques and the application of noninvasive and clinically approved imaging techniques to monitor delivery and therapeutic responses. Importantly, we demonstrate unprecedented rapid anti-inflammatory effects in atherosclerotic lesions after the nanomedical therapy.


The Lancet | 2017

Relation between resting amygdalar activity and cardiovascular events: a longitudinal and cohort study

Ahmed Tawakol; Amorina Ishai; Richard A. P. Takx; Amparo L. Figueroa; Abdelrahman Ali; Yannick Kaiser; Quynh A. Truong; Chloe Je Solomon; Claudia Calcagno; Venkatesh Mani; Cheuk Y. Tang; Willem J. M. Mulder; James W. Murrough; Udo Hoffmann; Matthias Nahrendorf; Lisa M. Shin; Zahi A. Fayad; Roger K. Pitman

BACKGROUND Emotional stress is associated with increased risk of cardiovascular disease. We imaged the amygdala, a brain region involved in stress, to determine whether its resting metabolic activity predicts risk of subsequent cardiovascular events. METHODS Individuals aged 30 years or older without known cardiovascular disease or active cancer disorders, who underwent 18F-fluorodexoyglucose PET/CT at Massachusetts General Hospital (Boston, MA, USA) between Jan 1, 2005, and Dec 31, 2008, were studied longitudinally. Amygdalar activity, bone-marrow activity, and arterial inflammation were assessed with validated methods. In a separate cross-sectional study we analysed the relation between perceived stress, amygdalar activity, arterial inflammation, and C-reactive protein. Image analyses and cardiovascular disease event adjudication were done by mutually blinded researchers. Relations between amygdalar activity and cardiovascular disease events were assessed with Cox models, log-rank tests, and mediation (path) analyses. FINDINGS 293 patients (median age 55 years [IQR 45·0-65·5]) were included in the longitudinal study, 22 of whom had a cardiovascular disease event during median follow-up of 3·7 years (IQR 2·7-4·8). Amygdalar activity was associated with increased bone-marrow activity (r=0·47; p<0·0001), arterial inflammation (r=0·49; p<0·0001), and risk of cardiovascular disease events (standardised hazard ratio 1·59, 95% CI 1·27-1·98; p<0·0001), a finding that remained significant after multivariate adjustments. The association between amygdalar activity and cardiovascular disease events seemed to be mediated by increased bone-marrow activity and arterial inflammation in series. In the separate cross-sectional study of patients who underwent psychometric analysis (n=13), amygdalar activity was significantly associated with arterial inflammation (r=0·70; p=0·0083). Perceived stress was associated with amygdalar activity (r=0·56; p=0·0485), arterial inflammation (r=0·59; p=0·0345), and C-reactive protein (r=0·83; p=0·0210). INTERPRETATION In this first study to link regional brain activity to subsequent cardiovascular disease, amygdalar activity independently and robustly predicted cardiovascular disease events. Amygdalar activity is involved partly via a path that includes increased bone-marrow activity and arterial inflammation. These findings provide novel insights into the mechanism through which emotional stressors can lead to cardiovascular disease in human beings. FUNDING None.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Probing nanoparticle translocation across the permeable endothelium in experimental atherosclerosis

YongTae Kim; Mark E. Lobatto; Tomohiro Kawahara; Bomy Lee Chung; Aneta J. Mieszawska; Brenda L. Sanchez-Gaytan; Francois Fay; Max L. Senders; Claudia Calcagno; Jacob R. Becraft; May Tun Saung; Ronald E. Gordon; Erik S.G. Stroes; Mingming Ma; Omid C. Farokhzad; Zahi A. Fayad; Willem J. M. Mulder; Robert Langer

Significance This study shows that an endothelialized microfluidic chip with controllable permeability can serve as a model for nanoparticle translocation across the permeable endothelium. Integration of this in vitro model and an in vivo rabbit model revealed that the extravasation of nanoparticles across the endothelium in atherosclerotic plaques depends on microvascular permeability. This approach represents a unique method for the assessment of nanoparticle behavior across the atherosclerotic endothelium, and may also serve as a valuable tool to study nanomedicine accumulation in a variety of other diseases. 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.


Contrast Media & Molecular Imaging | 2010

A fluorescent, paramagnetic and PEGylated gold/silica nanoparticle for MRI, CT and fluorescence imaging

Matti M. van Schooneveld; David P. Cormode; Rolf Koole; J. Timon van Wijngaarden; Claudia Calcagno; Torjus Skajaa; Jan Hilhorst; Dannis C. 't Hart; Zahi A. Fayad; Willem J. M. Mulder; Andries Meijerink

An important challenge in medical diagnostics is to design all-in-one contrast agents that can be detected with multiple techniques such as magnetic resonance imaging (MRI), X-ray computed tomography (CT), positron emission tomography (PET), single photon emission tomography (SPECT) or fluorescence imaging (FI). Although many dual labeled agents have been proposed, mainly for combined MRI/FI, constructs for three imaging modalities are scarce. Here gold/silica nanoparticles with a poly(ethylene glycol), paramagnetic and fluorescent lipid coating were synthesized, characterized and applied as trimodal contrast agents to allow for nanoparticle-enhanced imaging of macrophage cells in vitro via MRI, CT and FI, and mice livers in vivo via MRI and CT. This agent can be a useful tool in a multitude of applications, including cell tracking and target-specific molecular imaging, and is a step in the direction of truly multi-modal imaging.


Jacc-cardiovascular Imaging | 2011

Pioglitazone modulates vascular inflammation in atherosclerotic rabbits : noninvasive assessment with FDG-PET-CT and dynamic contrast-enhanced MR imaging

Esad Vucic; Stephen D. Dickson; Claudia Calcagno; James H.F. Rudd; Erin Moshier; Katsumi Hayashi; Jessica S. Mounessa; Michelle Roytman; Matthew J. Moon; James Lin; Sotirios Tsimikas; Edward A. Fisher; Klaas Nicolay; Valentin Fuster; Zahi A. Fayad

OBJECTIVES We sought to determine the antiatherosclerotic properties of pioglitazone using multimethod noninvasive imaging techniques. BACKGROUND Inflammation is an essential component of vulnerable or high-risk atheromas. Pioglitazone, a peroxisome proliferator-activated receptor-gamma agonist, possesses potent anti-inflammatory properties. We aimed to quantify noninvasively the anti-inflammatory effects of pioglitazone on atheroma using (18)F-fluorodeoxyglucose ((18)F-FDG) positron emission tomography (PET)/computed tomography (CT) and dynamic contrast-enhanced (DCE) magnetic resonance imaging (MRI). METHODS Atherosclerotic plaques were induced in the aorta of 15 New Zealand white rabbits by a combination of a hyperlipidemic diet and 2 balloon endothelial denudations. Nine rabbits continued the same diet, whereas 6 rabbits received pioglitazone (10 mg/kg orally) in addition to the diet. Twelve animals underwent (18)F-FDG-PET/CT, and 15 animals underwent DCE-MRI at baseline, 1 month, and 3 months after treatment initiation. Concomitantly, serum metabolic parameters were monitored. After imaging was completed, aortic histologic analysis and correlation analysis were performed. RESULTS The (18)F-FDG-PET/CT imaging detected an increase in average standardized uptake value in the control group (p < 0.01), indicating progressive inflammation, whereas stable standardized uptake values were observed in the treatment group, indicating no progression. The DCE-MRI analysis detected a significant decrease in the area under the curve for the pioglitazone group (p < 0.01). Immunohistologic examination of the aortas demonstrated a significant decrease in macrophage and oxidized phospholipid immunoreactivity in the pioglitazone group (p = 0.04 and p = 0.01, respectively) with respect to control animals, underlining the imaging results. Serum metabolic parameters showed no difference between groups. Strong positive correlations between standardized uptake value and macrophage density and between area under the curve and neovessels were detected (r(2) = 0.86 and p < 0.0001, and r(2) = 0.66 and p = 0.004, respectively). CONCLUSIONS Both (18)F-FDG-PET/CT and DCE-MRI demonstrate noninvasively the anti-inflammatory effects of pioglitazone on atheroma. Both imaging methods seem suited to monitor inflammation in atherosclerosis.


Science Advances | 2015

Inhibiting macrophage proliferation suppresses atherosclerotic plaque inflammation.

Jun Tang; Mark E. Lobatto; Laurien Hassing; Susanne E. M. van der Staay; Sarian M. van Rijs; Claudia Calcagno; Mounia S. Braza; Samantha Baxter; Francois Fay; Brenda L. Sanchez-Gaytan; Raphaël Duivenvoorden; Hendrik B. Sager; Yaritzy M Astudillo; Wei Sin Leong; Gert Storm; Carlos Pérez-Medina; Thomas Reiner; David P. Cormode; Gustav J. Strijkers; Erik S.G. Stroes; Filip K. Swirski; Matthias Nahrendorf; Edward A. Fisher; Zahi A. Fayad; Willem J. M. Mulder

Nanoparticle-based delivery of simvastatin inhibits plaque macrophage proliferation in apolipoprotein E–deficient mice. 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 the rapid reduction of plaque inflammation and favorable phenotype remodeling. We then combined this short-term nanoparticle intervention with an 8-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.


Bioconjugate Chemistry | 2009

Comparison of synthetic high density lipoprotein (HDL) contrast agents for MR imaging of atherosclerosis.

David P. Cormode; Rohith Chandrasekar; Amanda Delshad; Karen C. Briley-Saebo; Claudia Calcagno; Alessandra Barazza; Willem J. M. Mulder; Edward A. Fisher; Zahi A. Fayad

Determining arterial macrophage expression is an important goal in the molecular imaging of atherosclerosis. Here, we compare the efficacy of two synthetic, high density lipoprotein (HDL) based contrast agents for magnetic resonance imaging (MRI) of macrophage burden. Each form of HDL was labeled with gadolinium and rhodamine to allow MRI and fluorescence microscopy. Either the 37 or 18 amino acid peptide replaced the apolipoprotein A-I in these agents, which were termed 37pA-Gd or 18A-Gd. The diameters of 37pA-Gd and 18A-Gd are 7.6 and 8.0 nm, respectively, while the longitudinal relaxivities are 9.8 and 10.0 (mM s)(-1). 37pA has better lipid binding properties. In vitro tests with J774A.1 macrophages proved the particles possessed the functionality of HDL by eliciting cholesterol efflux and were taken up in a receptor-like fashion by the cells. Both agents produced enhancements in atherosclerotic plaques of apolipoprotein E knockout mice of approximately 90% (n = 7 per agent) and are macrophage specific as evidenced by confocal microscopy on aortic sections. The half-lives of 37pA-Gd and 18A-Gd are 2.6 and 2.1 h, respectively. Despite the more favorable lipid interactions of 37pA, both agents gave similar, excellent contrast for the detection of atherosclerotic macrophages using MRI.


Jacc-cardiovascular Imaging | 2012

Regression of Inflammation in Atherosclerosis by the LXR Agonist R211945 A Noninvasive Assessment and Comparison With Atorvastatin

Esad Vucic; Claudia Calcagno; Stephen D. Dickson; James H.F. Rudd; Katsumi Hayashi; Jan Bucerius; Erin Moshier; Jessica S. Mounessa; Michelle Roytman; Matthew J. Moon; James Lin; Tatsuo Tanimoto; Karen Brown; Masakatsu Kotsuma; Sotirios Tsimikas; Edward A. Fisher; Klaas Nicolay; Valentin Fuster; Zahi A. Fayad

OBJECTIVES The aim of this study was to noninvasively detect the anti-inflammatory properties of the novel liver X receptor agonist R211945. BACKGROUND R211945 induces reversal cholesterol transport and modulates inflammation in atherosclerotic plaques. We aimed to characterize with (18)F-fluorodeoxyglucose (FDG)-positron emission tomography (PET)/computed tomography (CT) and dynamic contrast-enhanced cardiac magnetic resonance (DCE-CMR) inflammation and neovascularization, respectively, in atherosclerotic plaques with R211945 treatment compared with atorvastatin treatment and a control. METHODS Twenty-one atherosclerotic New Zealand white rabbits were divided into 3 groups (control, R211945 [3 mg/kg orally], and atorvastatin [3 mg/kg orally] groups). All groups underwent (18)F-FDG-PET/CT and DCE-CMR at baseline and at 1 and 3 months after treatment initiation. Concomitantly, serum metabolic parameters and histology were assessed. For statistical analysis, continuous DCE-CMR and PET/CT outcomes were modeled as linear functions of time by using a linear mixed model, whereas the histological data, animal characteristics data, and nonlinear regression imaging data were analyzed with a 2-tailed Student t test. RESULTS (18)F-FDG-PET/CT detected a decrease in mean and maximum standard uptake values (SUV) over time in the R211945 group (both p = 0.001), indicating inflammation regression. The atorvastatin group displayed no significant change (p = 0.371 and p = 0.600, respectively), indicating no progression or regression. The control group demonstrated an increase in SUV (p = 0.01 and p = 0.04, respectively), indicating progression. There was a significant interaction between time and group for mean and maximum SUV (p = 0.0003 and p = 0.0016, respectively) . DCE-CMR detected a trend toward difference (p = 0.06) in the area under the curve in the atorvastatin group, suggesting a decrease in neovascularization. There was no significant interaction between time and group (p = 0.6350 and p = 0.8011, respectively). Macrophage and apolipoprotein B immunoreactivity decreased in the R211945 and atorvastatin groups (p < 0.0001 and p = 0.0004, respectively), and R211945 decreased oxidized phospholipid immunoreactivity (p = 0.02). CONCLUSIONS Noninvasive imaging with (18)F-FDG-PET/CT and DCE-CMR and histological analysis demonstrated significant anti-inflammatory effects of the LXR agonist R211945 compared with atorvastatin. The results suggest a possible role for LXR agonists in the treatment of atherosclerosis.

Collaboration


Dive into the Claudia Calcagno's collaboration.

Top Co-Authors

Avatar

Zahi A. Fayad

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Willem J. M. Mulder

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Venkatesh Mani

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Philip M. Robson

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Valentin Fuster

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Francois Fay

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Mark E. Lobatto

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Carlos Pérez-Medina

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Max L. Senders

Icahn School of Medicine at Mount Sinai

View shared research outputs
Researchain Logo
Decentralizing Knowledge