Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Drug Target ; 31(1): 109-118, 2023 01.
Article in English | MEDLINE | ID: mdl-35938912

ABSTRACT

Peri-stent restenosis following stent implantation is a major clinical problem. We have previously demonstrated that ultrasound-facilitated liposomal delivery of pioglitazone (PGN) to the arterial wall attenuated in-stent restenosis. To evaluate ultrasound mediated arterial delivery, in Yucatan miniswine, balloon inflations were performed in the carotid and subclavian arteries to simulate stent implantation and induce fibrin formation. The fibrin-binding peptide, GPRPPGGGC, was conjugated to echogenic liposomes (ELIP) containing dinitrophenyl-L-alanine-labelled pioglitazone (DNP-PGN) for targeting purposes. After pre-treating the arteries with nitroglycerine, fibrin-binding peptide-conjugated PGN-loaded ELIP (PAFb-DNP-PGN-ELIP also termed atheroglitatide) were delivered to the injured arteries via an endovascular catheter with an ultrasound core, either with or without ultrasound application (EKOSTM Endovascular System, Boston Scientific). In arteries treated with atheroglitatide, there was substantial delivery of PGN into the superficial layers (5 µm from the lumen) of the arteries with and without ultrasound, [(1951.17 relative fluorescence units (RFU) vs. 1901.17 RFU; P-value = 0.939)]. With ultrasound activation there was increased penetration of PGN into the deeper arterial layers (up to 35 µm from the lumen) [(13195.25 RFU vs. 7681.00 RFU; P-value = 0.005)]. These pre-clinical data demonstrate ultrasound mediated therapeutic vascular delivery to deeper layers of the injured arterial wall. This model has the potential to reduce peri- stent restenosis.


Subject(s)
Arteries , Liposomes , Pioglitazone , Ultrasonography , Stents
2.
JACC Basic Transl Sci ; 5(1): 1-11, 2020 Jan.
Article in English | MEDLINE | ID: mdl-32043017

ABSTRACT

Late in-stent restenosis remains a significant problem. Bare-metal stents were implanted into peripheral arteries in miniature swine, followed by direct intra-arterial infusion of nitric oxide-loaded echogenic liposomes (ELIPs) and anti-intercellular adhesion molecule-1 conjugated ELIPs loaded with pioglitazone exposed to an endovascular catheter with an ultrasonic core. Ultrasound-facilitated delivery of ELIP formulations into stented peripheral arteries attenuated neointimal growth. Local atheroma-targeted, ultrasound-triggered delivery of nitric oxide and pioglitazone, an anti-inflammatory peroxisome proliferator-activated receptor-γ agonist, into stented arteries has the potential to stabilize stent-induced neointimal growth and obviate the need for long-term antiplatelet therapy.

3.
Nanomedicine ; 14(6): 1941-1947, 2018 08.
Article in English | MEDLINE | ID: mdl-29933021

ABSTRACT

Management of patients suffering from myocardial infarction (MI) is based on the extent of coronary artery disease and myocardial scar burden. We have developed a potentially clinically-useful X-ray molecular imaging contrast agent based on gold nanoparticle (AuNPs) functionalized with collagen-binding adhesion protein 35 (CNA35) with the capabilities of achieving prolonged blood pool enhancement for vascular imaging of the coronary arteries and specific targeting of collagen within myocardial scar. At a concentration of ~ 45 mg Au/ml, AuNPs maintained a stable blood pool enhancement at 142-160 HU within an hour of intravenous administration. At 6 hours, specific signal enhancement was detected in the myocardium scar in rats injected with CNA35-AuNPs, but not with control AuNPs or in control animals. In conclusion, CNA35-AuNPs may be considered as a CT contrast agent for both vascular imaging of coronary artery disease and molecular imaging of myocardial scar in the heart.


Subject(s)
Cell Adhesion Molecules/metabolism , Cicatrix/pathology , Gold/chemistry , Metal Nanoparticles/administration & dosage , Myocardial Infarction/pathology , Myocardium/pathology , Tomography, X-Ray Computed/methods , Animals , Cell Adhesion , Cell Adhesion Molecules/chemistry , Cicatrix/diagnostic imaging , Female , Image Processing, Computer-Assisted , Metal Nanoparticles/chemistry , Myocardial Infarction/diagnostic imaging , Rats
4.
Ultrasound Med Biol ; 41(6): 1701-10, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25819469

ABSTRACT

The aim of this study was to determine whether pre-treatment with nitric oxide-loaded echogenic liposomes (NO-ELIP) plus ultrasound can improve highlighting by molecularly targeted (anti-vascular cell adhesion molecule 1 [VCAM-1]) ELIP of atheroma components. Atherosclerotic animals were treated with anti-VCAM-1-ELIP or immunoglobulin (IgG)-ELIP. Each group was selected at random to receive pre-treatment with standard ELIP plus ultrasound, NO-ELIP without ultrasound and NO-ELIP plus ultrasound. Intravascular ultrasound highlighting data for the same arterial segments were collected before and after treatment. Pre-treatment with NO-ELIP plus ultrasound resulted in a significant increase in acoustic enhancement by anti-VCAM-1-ELIP (21.3 ± 1.5% for gray-scale value, 53.9 ± 3.1% for radiofrequency data; p < 0.001 vs. IgG-ELIP, p < 0.05 vs. pre-treatment with standard ELIP plus ultrasound or NO-ELIP without ultrasound). NO-ELIP plus ultrasound can improve highlighting of atheroma by anti-VCAM-1 ELIP. This NO pre-treatment strategy may be useful in optimizing contrast agent delivery to the vascular wall for both diagnostic and therapeutic applications.


Subject(s)
Liposomes/metabolism , Molecular Imaging/methods , Nitric Oxide/metabolism , Plaque, Atherosclerotic/diagnostic imaging , Vascular Cell Adhesion Molecule-1/metabolism , Animals , Disease Models, Animal , Plaque, Atherosclerotic/metabolism , Swine , Swine, Miniature , Ultrasonography
5.
Ultrasound Med Biol ; 40(6): 1167-76, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24613216

ABSTRACT

We present an ultrasound technique for the detection of inflammatory changes in developing atheromas. We used contrast-enhanced ultrasound imaging with (i) microbubbles targeted to intercellular adhesion molecule-1 (ICAM-1), a molecule of adhesion involved in inflammatory processes in lesions of atheromas in New Zealand White rabbits, and (ii) pretreatment with nitric oxide-loaded microbubbles and ultrasound activation at the site of the endothelium to enhance the permeability of the arterial wall and the penetration of ICAM-1-targeted microbubbles. This procedure increases acoustic enhancement 1.2-fold. Pretreatment with nitric oxide-loaded echogenic liposomes and ultrasound activation can potentially facilitate the subsequent penetration of targeted echogenic liposomes into the arterial wall, thus allowing improved detection of inflammatory changes in developing atheromas.


Subject(s)
Contrast Media/pharmacokinetics , Endothelium, Vascular/drug effects , Endothelium, Vascular/diagnostic imaging , Intercellular Adhesion Molecule-1/metabolism , Liposomes/pharmacokinetics , Nitric Oxide/pharmacology , Plaque, Atherosclerotic/diagnostic imaging , Animals , Disease Models, Animal , Microbubbles , Permeability/drug effects , Plaque, Atherosclerotic/metabolism , Rabbits , Ultrasonography
6.
Atherosclerosis ; 231(2): 252-60, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24267236

ABSTRACT

OBJECTIVE: This study aimed to demonstrate whether pretreatment with nitric oxide (NO) loaded into echogenic immunoliposomes (ELIP) plus ultrasound, applied before injection of molecularly targeted ELIP can promote penetration of the targeted contrast agent and improve visualization of atheroma components. METHODS: ELIP were prepared using the pressurization-freeze method. Atherosclerosis was induced in Yucatan miniswine by balloon denudation and a hyperlipidemic diet. The animals were randomized to receive anti-intercellular adhesion molecule-1 (ICAM-1) ELIP or immunoglobulin (IgG)-ELIP, and were subdivided to receive pretreatment with standard ELIP plus ultrasound, NO-loaded ELIP, or NO-loaded ELIP plus ultrasound. Intravascular ultrasound (IVUS) data were collected before and after treatment. RESULTS: Pretreatment with standard ELIP plus ultrasound or NO-loaded ELIP without ultrasound resulted in 9.2 ± 0.7% and 9.2 ± 0.8% increase in mean gray scale values, respectively, compared to baseline (p < 0.001 vs. control). Pretreatment with NO-loaded ELIP plus ultrasound activation resulted in a further increase in highlighting with a change in mean gray scale value to 14.7 ± 1.0% compared to baseline (p < 0.001 vs. control). These differences were best appreciated when acoustic backscatter data values (RF signal) were used [22.7 ± 2.0% and 22.4 ± 2.2% increase in RF signals for pretreatment with standard ELIP plus ultrasound and NO-loaded ELIP without ultrasound respectively (p < 0.001 vs. control), and 40.0 ± 2.9% increase in RF signal for pretreatment with NO-loaded ELIP plus ultrasound (p < 0.001 vs. control)]. CONCLUSION: NO-loaded ELIP plus ultrasound activation can facilitate anti-ICAM-1 conjugated ELIP delivery to inflammatory components in the arterial wall. This NO pretreatment strategy has potential to improve targeted molecular imaging of atheroma for eventual true tailored and personalized management of cardiovascular diseases.


Subject(s)
Liposomes/chemistry , Molecular Imaging/methods , Nitric Oxide/chemistry , Plaque, Atherosclerotic/diagnosis , Acoustics , Animals , Hyperlipidemias , Intercellular Adhesion Molecule-1/metabolism , Molecular Imaging/instrumentation , Permeability , Plaque, Atherosclerotic/diagnostic imaging , Plaque, Atherosclerotic/genetics , Random Allocation , Swine , Swine, Miniature , Ultrasonics , Ultrasonography
7.
Circulation ; 122(16): 1578-87, 2010 Oct 19.
Article in English | MEDLINE | ID: mdl-20921443

ABSTRACT

BACKGROUND: Ischemia-related neurological injury is a primary cause of stroke disability. Studies have demonstrated that xenon (Xe) may have potential as an effective and nontoxic neuroprotectant. Xe delivery is, however, hampered by lack of suitable administration methods. We have developed a pressurization-freeze method to encapsulate Xe into echogenic liposomes (Xe-ELIP) and have modulated local gas release with transvascular ultrasound exposure. METHODS AND RESULTS: Fifteen microliters of Xe were encapsulated into each 1 mg of liposomes (70% Xe and 30% argon). Xe delivery from Xe-ELIP into cells and consequent neuroprotective effects were evaluated with oxygen/glucose-deprived and control neuronal cells in vitro. Xe-ELIP were administered into Sprague-Dawley rats intravenously or intra-arterially after right middle cerebral artery occlusion. One-megahertz low-amplitude (0.18 MPa) continuous wave ultrasound directed onto the internal carotid artery triggered Xe release from circulating Xe-ELIP. Effects of Xe delivery on ischemia-induced neurological injury and disability were evaluated. Xe-ELIP delivery to oxygen/glucose-deprived neuronal cells improved cell viability in vitro and resulted in a 48% infarct volume decrease in vivo. Intravenous Xe-ELIP administration in combination with the ultrasound directed onto the carotid artery enhanced local Xe release from circulating Xe-ELIP and demonstrated 75% infarct volume reduction. This was comparable to the effect after intra-arterial administration. Behavioral tests on limb placement and grid and beam walking correlated with infarct reduction. CONCLUSIONS: This novel methodology may provide a noninvasive strategy for ultrasound-enhanced local therapeutic gas delivery for cerebral ischemia-related injury while minimizing systemic side effects.


Subject(s)
Brain Ischemia/prevention & control , Drug Delivery Systems/methods , Infarction, Middle Cerebral Artery/complications , Reperfusion Injury/prevention & control , Xenon/administration & dosage , Animals , Brain Ischemia/etiology , Cell Survival/physiology , Injections, Intravenous , Liposomes , Male , Models, Animal , Neurons/cytology , Neurons/physiology , Rats , Rats, Sprague-Dawley , Reperfusion Injury/etiology , Ultrasonography
8.
Invest Radiol ; 45(10): 685-91, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20733507

ABSTRACT

OBJECTIVES: This study aimed to demonstrate three-dimensional (3D) visualization of early/inflammatory arterial atheroma using intravascular ultrasound (IVUS) and targeted echogenic immunoliposomes (ELIP). IVUS can be used as a molecular imaging modality with the use of targeted contrast agents for atheroma detection. Three-dimensional reconstruction of 2-dimensional IVUS images may provide improved atheroma visualization. MATERIALS AND METHODS: Atheroma were induced in arteries of Yucatan miniswine (n = 5) by endothelial cell denudation followed by a 4-week high cholesterol diet. The contralateral arteries were left intact and served as controls. Anti-intercellular adhesion molecule-1 (ICAM-1) and generic gammaglobulin (IgG) conjugated ELIP were prepared. Arteries were imaged using IVUS before and after ELIP injection. Images were digitized, manually traced, segmented, and placed in tomographic sequence for 3D visualization. Atheroma brightness enhancement was compared and reported as mean gray scale values. Plaque volume was quantified both from IVUS and histologic images. RESULTS: Anti-ICAM-1 ELIP highlighting of the atheroma in all arterial segments was different compared with baseline (P < 0.05). There was no difference in the mean gray scale values with IgG-ELIP. Arterial 3D IVUS images allowed visualization of the entire plaque distribution. The highlighted plaque/atheroma volume with anti-ICAM-1 ELIP was greater than baseline (P < 0.01). CONCLUSION: This study demonstrates specific highlighting of early/inflammatory atheroma in vivo using anti-ICAM-1 ELIP. Three-dimensional IVUS reconstruction provides good visualization of plaque distribution in the arterial wall. This novel methodology may help to detect and diagnose pathophysiologic development of all stages of atheroma formation in vivo and quantitate plaque volume for serial and long-term atherosclerotic treatment studies.


Subject(s)
Endothelium, Vascular/diagnostic imaging , Liposomes , Plaque, Atherosclerotic/diagnostic imaging , Ultrasonography, Interventional , Analysis of Variance , Animals , Echocardiography, Three-Dimensional , Endothelium, Vascular/pathology , Enzyme-Linked Immunosorbent Assay , Immunoglobulin G , In Vitro Techniques , Intercellular Adhesion Molecule-1 , Molecular Imaging , Plaque, Atherosclerotic/pathology
9.
J Am Coll Cardiol ; 54(7): 652-9, 2009 Aug 11.
Article in English | MEDLINE | ID: mdl-19660697

ABSTRACT

OBJECTIVES: We sought to develop a new bioactive gas-delivery method by the use of echogenic liposomes (ELIP) as the gas carrier. BACKGROUND: Nitric oxide (NO) is a bioactive gas with potent therapeutic effects. The bioavailability of NO by systemic delivery is low with potential systemic effects. METHODS: Liposomes containing phospholipids and cholesterol were prepared by the use of a new method, freezing under pressure. The encapsulation and release profile of NO from NO-containing ELIP (NO-ELIP) or a mixture of NO/argon (NO/Ar-ELIP) was studied. The uptake of NO from NO-ELIP by cultured vascular smooth muscle cells (VSMCs) both in the absence and presence of hemoglobin was determined. The effect of NO-ELIP delivery to attenuate intimal hyperplasia in a balloon-injured artery was determined. RESULTS: Coencapsulation of NO with Ar enabled us to adjust the amount of encapsulated NO. A total of 10 microl of gas can be encapsulated into 1 mg of liposomes. The release profile of NO from NO-ELIP demonstrated an initial rapid release followed by a slower release during the course of 8 h. Sixty-eight percent of cells remained viable when incubated with 80 microg/ml of NO/Ar-ELIP for 4 h. The delivery agent of NO to VSMCs by the use of NO/Ar-ELIP was 7-fold greater than unencapsulated NO. We discovered that NO/Ar-ELIP remained an effective delivery agent of NO to VSMCs even in the presence of hemoglobin. Local NO-ELIP administration to balloon-injured carotid arteries attenuated the development of intimal hyperplasia and reduced arterial wall thickening by 41 +/- 9%. CONCLUSIONS: Liposomes can protect and deliver a bioactive gas to target tissues with the potential for both visualization of gas delivery and controlled therapeutic gas release.


Subject(s)
Drug Delivery Systems , Liposomes , Muscle, Smooth, Vascular/pathology , Nitric Oxide/administration & dosage , Tunica Intima/pathology , Animals , Biological Availability , Hyperplasia/prevention & control , Muscle, Smooth, Vascular/cytology , Rats , Tunica Intima/drug effects , Ultrasonics
10.
J Ultrasound Med ; 27(11): 1597-606, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18946099

ABSTRACT

OBJECTIVE: To achieve ultrasound-controlled drug delivery using echogenic liposomes (ELIPs), we assessed ultrasound-triggered release of hydrophilic and lipophilic agents in vitro using color Doppler ultrasound delivered with a clinical 6-MHz compact linear array transducer. METHODS: Calcein, a hydrophilic agent, and papaverine, a lipophilic agent, were each separately loaded into ELIPs. Calcein-loaded ELIP (C-ELIP) and papaverine-loaded ELIP (P-ELIP) solutions were circulated in a flow model and treated with 6-MHz color Doppler ultrasound or Triton X-100. Treatment with Triton X-100 was used to release the encapsulated calcein or papaverine content completely. The free calcein concentration in the solution was measured directly by spectrofluorimetry. The free papaverine in the solution was separated from liposome-bound papaverine by spin column filtration, and the resulting papaverine concentration was measured directly by absorbance spectrophotometry. Dynamic changes in echogenicity were assessed with low-output B-mode ultrasound (mechanical index, 0.04) as mean digital intensity. RESULTS: Color Doppler ultrasound caused calcein release from C-ELIPs compared with flow alone (P < .05) but did not induce papaverine release from P-ELIPs compared with flow alone (P > .05). Triton X-100 completely released liposome-associated calcein and papaverine. Initial echogenicity was higher for C-ELIPs than P-ELIPs. Color Doppler ultrasound and Triton X-100 treatments reduced echogenicity for both C-ELIPs and P-ELIPs (P < .05). CONCLUSIONS: The differential efficiency of ultrasound-mediated pharmaceutical release from ELIPs for water- and lipid-soluble compounds suggests that water-soluble drugs are better candidates for the design and development of ELIP-based ultrasound-controlled drug delivery systems.


Subject(s)
Drug Delivery Systems/methods , Fluoresceins/chemistry , Liposomes/chemistry , Liposomes/radiation effects , Papaverine/chemistry , Sonication , Diffusion , Fluoresceins/administration & dosage , Hydrophobic and Hydrophilic Interactions , Papaverine/administration & dosage , Phosphodiesterase Inhibitors/administration & dosage , Phosphodiesterase Inhibitors/chemistry
11.
J Liposome Res ; 18(4): 263-77, 2008.
Article in English | MEDLINE | ID: mdl-18720194

ABSTRACT

BACKGROUND: development of encapsulated therapeutics that could be released upon ultrasound exposure has strong implications for enhancing drug effects at the target site. We have developed echogenic liposomes (ELIP) suitable for ultrasound imaging of blood flow and ultrasound-mediated intravascular drug release. Papaverine was chosen as the test drug because its clinical application requires high concentration in the target vascular bed but low concentration in the systemic circulation. METHODS: the procedure for preparation of standard ELIP was modified by including Papaverine hydrochloride in the lipid hydration solution, followed by three freeze-thaw cycles to increase encapsulation of the drug. Sizing and encapsulation pharmacokinetics were performed using a Coulter counter and a phosphodiesterase activity assay. Stability of Papaverine-loaded ELIP (PELIP) was monitored with a clinical diagnostic ultrasound scanner equipped with a linear array transducer at a center frequency of 4.5 MHz by assessing the mean digital intensity within a region of interest over time. The stability of PELIP was compared to those of standard ELIP and Optison. RESULTS: relative to standard ELIP, PELIP were larger (median diameter = 1.88 +/- 0.10 microm for PELIP vs 1.08 +/- 0.15 microm for ELIP) and had lower Mean Gray Scale Values (MGSV) (92 +/- 24.8 for PELIP compared to 142.3 +/- 10.7 for ELIP at lipid concentrations of 50 microg/ml). The maximum loading efficiency and mean encapsulated concentration were 24% +/- 7% and 2.1 +/- 0.7 mg/ml, respectively. Papaverine retained its phosphodiesterase inhibitory activity when associated with PELIP. Furthermore, a fraction of this activity remained latent until released by dissolution of liposomal membranes with detergent. The stability of both PELIP and standard ELIP were similar, but both are greater than that of Optison. CONCLUSIONS: our results suggest that PELIP have desirable physical, biochemical, biological, and acoustic characteristics for potential in vivo administration and ultrasound-controlled drug delivery.


Subject(s)
Drug Delivery Systems , Liposomes , Papaverine , Ultrasonics , Acoustics , Animals , Cattle , Drug Compounding , Liposomes/chemistry , Liposomes/metabolism , Papaverine/chemistry , Papaverine/metabolism , Particle Size , Phosphodiesterase Inhibitors/chemistry , Phosphodiesterase Inhibitors/metabolism , Phosphoric Diester Hydrolases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL