Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Chem Sci ; 8(9): 6176-6181, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28989649

ABSTRACT

Photoacoustic (PA) imaging is an emerging non-invasive diagnostic modality with many potential clinical applications in oncology, rheumatology and the cardiovascular field. For this purpose, there is a high demand for exogenous contrast agents with high absorption coefficients in the optical window for tissue imaging, i.e. the near infrared (NIR) range between 680 and 950 nm. We herein report the photoacoustic properties of quinone-fused porphyrins inserted with different transition metals as new highly promising candidates. These dyes exhibit intense NIR absorption, a lack of fluorescence emission, and PA sensitivity in concentrations below 3 nmol mL-1. In this context, the highest PA signal was obtained with a Zn(ii) inserted dye. Furthermore, this dye was stable in blood serum and free thiol solution and exhibited negligible cell toxicity. Additionally, the Zn(ii) probe could be detected with an up to 3.2 fold higher PA intensity compared to the clinically most commonly used PA agent, ICG. Thus, further exploration of the 'quinone-fusing' approach to other chromophores may be an efficient way to generate highly potent PA agents that do not fluoresce and shift their absorption into the NIR range.

2.
J Control Release ; 259: 128-135, 2017 08 10.
Article in English | MEDLINE | ID: mdl-28279799

ABSTRACT

Microbubbles (MB) are routinely used as contrast agents for ultrasound (US) imaging. We describe different types of targeted and drug-loaded poly(n-butyl cyanoacrylate) (PBCA) MB, and demonstrate their suitability for multiple biomedical applications, including molecular US imaging and US-mediated drug delivery. Molecular imaging of angiogenic tumor blood vessels and inflamed atherosclerotic endothelium is performed by modifying the surface of PBCA MB with peptides and antibodies recognizing E-selectin and VCAM-1. Stable and inertial cavitation of PBCA MB enables sonoporation and permeabilization of blood vessels in tumors and in the brain, which can be employed for direct and indirect drug delivery. Direct drug delivery is based on US-induced release of (model) drug molecules from the MB shell. Indirect drug delivery refers to US- and MB-mediated enhancement of extravasation and penetration of co-administered drugs and drug delivery systems. These findings are in line with recently reported pioneering proof-of-principle studies showing the usefulness of (phospholipid) MB for molecular US imaging and sonoporation-enhanced drug delivery in patients. They aim to exemplify the potential and the broad applicability of combining MB with US to improve disease diagnosis and therapy.


Subject(s)
Drug Delivery Systems , Enbucrilate/administration & dosage , Microbubbles , Animals , Antibodies/administration & dosage , Antibodies/chemistry , Biotin/administration & dosage , Biotin/chemistry , Brain/metabolism , Carotid Arteries/diagnostic imaging , Carotid Arteries/metabolism , Cell Line, Tumor , Dextrans/administration & dosage , Dextrans/chemistry , E-Selectin/immunology , Enbucrilate/chemistry , Fluorescein-5-isothiocyanate/administration & dosage , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/chemistry , Fluorescent Dyes/administration & dosage , Fluorescent Dyes/chemistry , Humans , Male , Mice, Nude , Molecular Imaging , Neoplasms/diagnostic imaging , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic , Rhodamines/administration & dosage , Rhodamines/chemistry , Streptavidin/administration & dosage , Streptavidin/chemistry , Ultrasonic Waves , Ultrasonography , Vascular Cell Adhesion Molecule-1/immunology , Vascular Endothelial Growth Factor Receptor-2/immunology
3.
Invest Radiol ; 51(12): 767-775, 2016 12.
Article in English | MEDLINE | ID: mdl-27119438

ABSTRACT

OBJECTIVES: Interventions such as balloon angioplasty can cause vascular injury leading to platelet activation, thrombus formation, and inflammatory response. This induces vascular smooth muscle cell activation and subsequent re-endothelialization with expression of αvß3-integrin by endothelial cells and vascular smooth muscle cell. Thus, poly-N-butylcyanoacrylate microbubbles (MBs) targeted to αvß3-integrin were evaluated for monitoring vascular healing after vessel injury in pigs using molecular ultrasound imaging. MATERIALS AND METHODS: Approval for animal experiments was obtained. The binding specificity of αvß3-integrin-targeted MB to human umbilical vein endothelial cells was tested with fluorescence microscopy. In vivo imaging was performed using a clinical ultrasound system and an 8-MHz probe. Six mini pigs were examined after vessel injury in the left carotid artery. The right carotid served as control. Uncoated MB, cDRG-coated MB, and αvß3-integrin-specific cRGD-coated MB were injected sequentially. Bound MBs were assessed 8 minutes after injection using ultrasound replenishment analysis. Measurements were performed 2 hours, 1 and 5 weeks, and 3 and 6 months after injury. In vivo data were validated by immunohistochemistry. RESULTS: Significantly stronger binding of cRGD-MB than MB and cDRG-MB to human umbilical vein endothelial cells was found (P < 0.01). As vessel injury leads to upregulation of αvß3-integrin, cRGD-MBs bound significantly stronger (P < 0.05) in injured carotid arteries than at the counter side 1 week after vessel injury and significant differences could also be observed after 5 weeks. After 3 months, αvß3-integrin expression decreased to baseline and binding of cRGD-MB was comparable in both vessels. Values remained at baseline also after 6 months. CONCLUSIONS: Ultrasound imaging with RGD-MB is promising for monitoring vascular healing after vessel injury. This may open new perspectives to assess vascular damage after radiological interventions.


Subject(s)
Carotid Arteries/diagnostic imaging , Carotid Artery Injuries/diagnostic imaging , Integrin alphaVbeta3/metabolism , Ultrasonography/methods , Wound Healing/physiology , Animals , Carotid Arteries/metabolism , Carotid Artery Injuries/metabolism , Disease Models, Animal , Female , Humans , Immunohistochemistry , Microbubbles , Molecular Imaging/methods , Reproducibility of Results , Swine
4.
Mol Imaging Biol ; 18(2): 180-90, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26391990

ABSTRACT

PURPOSE: Our objective was to determine the lowest diagnostically effective dose for E-selectin-targeted poly n-butyl cyanoacrylate (PBCA)-shelled microbubbles and to apply it to monitor antiangiogenic therapy effects. PROCEDURES: PBCA-shelled microbubbles (MBs) coupled to an E-selectin-specific peptide were applied in mice carrying MLS or A431 carcinoma xenografts scaling down the MB dosage to the lowest level where binding could be examined with a 18-MHz small animal ultrasound transducer. Differences in E-selectin expression in the two carcinoma xenografts were confirmed by enzyme-linked immunosorbent assay (ELISA). In addition, MLS tumor-bearing mice under antiangiogenic therapy were monitored using E-selectin-targeted MBs at the lowest applicable dose. Therapy effects on tumor vascularization were verified by immunohistological analyses. RESULTS: The minimally required dosage was 7 × 10(7) MBs/kg body weight. This dosage was sufficient to enable E-selectin detection in high E-selectin-expressing MLS tumors, while low E-selectin-expressing A431 tumors required almost 2.5-fold higher doses. At the dose of 7 × 10(7) MBs/kg body weight, a decrease in E-selectin MB binding under antiangiogenic therapy could be assessed (being significant after 3 days of treatment; p < 0.0001), which was in line with the significant drop in E-selectin-positive area fractions that was found histologically (p < 0.05). CONCLUSIONS: Molecular ultrasound imaging with our E-selectin-targeted MB and therapy monitoring was possible down to a dose of 7 × 10(7) MBs/kg body weight (equates to 66 µg PBCA/kg and 4.6 mg PBCA/70 kg). Improvements in choice of targets, MB composition, and other MB detection methods may improve sensitivity and lead to reliable detection results of clinically transferrable MBs at even lower dosage levels.


Subject(s)
E-Selectin/metabolism , Enbucrilate/chemistry , Microbubbles , Molecular Imaging/methods , Ultrasonography/methods , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Fluorescent Antibody Technique , Humans , Mice, Nude , Neoplasms/blood supply , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Protein Binding , Tumor Burden , Xenograft Model Antitumor Assays
5.
Arterioscler Thromb Vasc Biol ; 35(6): 1366-73, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25838431

ABSTRACT

OBJECTIVE: Cardiovascular interventions induce damage to the vessel wall making antithrombotic therapy inevitable until complete endothelial recovery. Without a method to accurately determine the endothelial status, many patients undergo prolonged anticoagulation therapy, denying them any invasive medical procedures, such as surgical operations and dental interventions. Therefore, we aim to introduce molecular ultrasound imaging of the vascular cell adhesion molecule (VCAM)-1 using targeted poly-n-butylcyanoacrylate microbubbles (MB(VCAM-1)) as an easy accessible method to monitor accurately the reendothelialization of vessels. APPROACH AND RESULTS: ApoE(-/-) mice were fed with an atherogenic diet for 1 and 12 weeks and subsequently, endothelial denudation was performed in the carotid arteries using a guidewire. Molecular ultrasound imaging was performed at different time points after denudation (1, 3, 7, and 14 days). An increased MB(VCAM-1) binding after 1 day, a peak after 3 days, and a decrease after 7 days was found. After 12 weeks of diet, MB(VCAM-1) binding also peaked after 3 days but remained high until 7 days, indicating a delay in endothelial recovery. Two-photon laser scanning microscopy imaging of double fluorescence staining confirmed the exposure of VCAM-1 on the superficial layer after arterial injury only during the healing phase. After complete reendothelialization, VCAM-1 expression persisted in the subendothelial layer but was not reachable for the MBV(CAM-1) anymore. CONCLUSION: Molecular ultrasound imaging with MB(VCAM-1) is promising to assess vascular damage and to monitor endothelial recovery after arterial interventions. Thus, it may become an important diagnostic tool supporting the development of adequate therapeutic strategies to personalize anticoagulant and anti-inflammatory therapy after cardiovascular intervention.


Subject(s)
Atherosclerosis/surgery , Endothelium, Vascular/diagnostic imaging , Endothelium, Vascular/surgery , Vascular Cell Adhesion Molecule-1/metabolism , Animals , Atherosclerosis/diagnostic imaging , Biomarkers/metabolism , Carotid Arteries/diagnostic imaging , Carotid Arteries/pathology , Carotid Arteries/surgery , Disease Models, Animal , Enbucrilate , Endothelium, Vascular/injuries , Endothelium, Vascular/pathology , Endovascular Procedures , Mice , Microbubbles , Microscopy, Confocal , Ultrasonography , Wound Healing
6.
Adv Funct Mater ; 25(1): 36-43, 2015 Jan 07.
Article in English | MEDLINE | ID: mdl-25729344

ABSTRACT

Efficient and safe drug delivery across the blood-brain barrier (BBB) remains to be one of the major challenges of biomedical and (nano-) pharmaceutical research. Here, we show that poly(butyl cyanoacrylate)-based microbubbles (MB), carrying ultrasmall superparamagnetic iron oxide (USPIO) nanoparticles within their shell, can be used to mediate and monitor BBB permeation. Upon exposure to transcranial ultrasound pulses, USPIO-MB are destroyed, resulting in acoustic forces inducing vessel permeability. At the same time, USPIO are released from the MB shell, they extravasate across the permeabilized BBB and they accumulate in extravascular brain tissue, thereby providing non-invasive R2*-based magnetic resonance imaging information on the extent of BBB opening. Quantitative changes in R2* relaxometry were in good agreement with 2D and 3D microscopy results on the extravascular deposition of the macromolecular model drug FITC-dextran into the brain. Such theranostic materials and methods are considered to be useful for mediating and monitoring drug delivery across the BBB, and for enabling safe and efficient treatment of CNS disorders.

7.
Chem Commun (Camb) ; 51(28): 6084-7, 2015 Apr 11.
Article in English | MEDLINE | ID: mdl-25670068

ABSTRACT

Here we present a facile synthetic method yielding a linear form of polydopamine via Kumada-coupling, which can be converted into water-soluble melanin, generating high contrast in photoacoustic imaging.


Subject(s)
Indoles/chemistry , Melanins/pharmacokinetics , Photoacoustic Techniques , Polymers/chemistry , Water/chemistry , Animals , Chickens , Indoles/chemical synthesis , Injections, Subcutaneous , Melanins/administration & dosage , Melanins/chemistry , Mice , Molecular Structure , Polymers/chemical synthesis , Solubility
8.
J Control Release ; 182: 83-9, 2014 May 28.
Article in English | MEDLINE | ID: mdl-24631862

ABSTRACT

The Enhanced Permeability and Retention (EPR) effect is extensively used in drug delivery research. Taking into account that EPR is a highly variable phenomenon, we have here set out to evaluate if contrast-enhanced functional ultrasound (ceUS) imaging can be employed to characterize EPR-mediated passive drug targeting to tumors. Using standard fluorescence molecular tomography (FMT) and two different protocols for hybrid computed tomography-fluorescence molecular tomography (CT-FMT), the tumor accumulation of a ~10 nm-sized near-infrared-fluorophore-labeled polymeric drug carrier (pHPMA-Dy750) was evaluated in CT26 tumor-bearing mice. In the same set of animals, two different ceUS techniques (2D MIOT and 3D B-mode imaging) were employed to assess tumor vascularization. Subsequently, the degree of tumor vascularization was correlated with the degree of EPR-mediated drug targeting. Depending on the optical imaging protocol used, the tumor accumulation of the polymeric drug carrier ranged from 5 to 12% of the injected dose. The degree of tumor vascularization, determined using ceUS, varied from 4 to 11%. For both hybrid CT-FMT protocols, a good correlation between the degree of tumor vascularization and the degree of tumor accumulation was observed, within the case of reconstructed CT-FMT, correlation coefficients of ~0.8 and p-values of <0.02. These findings indicate that ceUS can be used to characterize and predict EPR, and potentially also to pre-select patients likely to respond to passively tumor-targeted nanomedicine treatments.


Subject(s)
Acrylamides/administration & dosage , Contrast Media/administration & dosage , Drug Delivery Systems , Neoplasms/diagnostic imaging , Neoplasms/metabolism , Animals , Blood Volume , Cell Line, Tumor , Enbucrilate , Mice, Nude , Microbubbles , Neoplasms/blood supply , Neoplasms/physiopathology , Permeability , Regional Blood Flow , Tomography/methods , Ultrasonography
9.
Adv Drug Deliv Rev ; 72: 15-27, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24316070

ABSTRACT

Ultrasound (US) imaging is an exquisite tool for the non-invasive and real-time diagnosis of many different diseases. In this context, US contrast agents can improve lesion delineation, characterization and therapy response evaluation. US contrast agents are usually micrometer-sized gas bubbles, stabilized with soft or hard shells. By conjugating antibodies to the microbubble (MB) surface, and by incorporating diagnostic agents, drugs or nucleic acids into or onto the MB shell, molecular, multimodal and theranostic MBs can be generated. We here summarize recent advances in molecular, multimodal and theranostic US imaging, and introduce concepts how such advanced MB can be generated, applied and imaged. Examples are given for their use to image and treat oncological, cardiovascular and neurological diseases. Furthermore, we discuss for which therapeutic entities incorporation into (or conjugation to) MB is meaningful, and how US-mediated MB destruction can increase their extravasation, penetration, internalization and efficacy.


Subject(s)
Ultrasonography/methods , Animals , Contrast Media , Drug Delivery Systems , Humans , Microbubbles , Ultrasonic Therapy
10.
Circ Cardiovasc Imaging ; 6(6): 974-81, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24036383

ABSTRACT

BACKGROUND: The ability to image incipient atherosclerosis is based on the early events taking place at the endothelial level. We hypothesized that the expression of intercellular adhesion molecule-1 even in vessels with high flow rates can be imaged at the molecular level using 2 complementary imaging techniques: 2-photon laser scanning microscopy and contrast-enhanced ultrasound. METHODS AND RESULTS: Using 2-photon laser scanning microscopy and contrast-enhanced ultrasound, intercellular adhesion molecule-1-targeted and rhodamine-loaded microbubbles were shown to be specifically bound to tumor necrosis factor-α-stimulated human umbilical vein endothelial cells and murine carotid arteries (44 wild-type mice) at shear stresses ranging from 1.25 to 120 dyn/cm(2). Intercellular adhesion molecule-1-targeted and rhodamine-loaded microbubbles bound 8× more efficient (P=0.016) to stimulated human umbilical vein endothelial cells than to unstimulated cells and 14× more than nontargeted microbubbles (P=0.016). In excised carotids, binding efficiency did not decrease significantly when increasing the flow rate from 0.25 to 0.6 mL/min. Higher flow rates (0.8 and 1 mL/min) showed significantly reduced microbubbles retention, by 38% (P=0.03) and 55% (P=0.03), respectively. Ex vivo results were translatable in vivo, confirming that intercellular adhesion molecule-1-targeted and rhodamine-loaded microbubbles are able to bind specifically to the inflamed carotid artery endothelia under physiological flow conditions and to be noninvasively detected using contrast-enhanced ultrasound. CONCLUSIONS: Our data provide groundwork for the implementation of molecular ultrasound imaging in vessels with high shear stress and flow rates, as well as for the future development of image-guided therapeutic interventions, and multiphoton microscopy as the appropriate method of validation.


Subject(s)
Atherosclerosis/diagnostic imaging , Contrast Media , Intercellular Adhesion Molecule-1 , Microbubbles , Molecular Imaging/methods , Rhodamines , Animals , Atherosclerosis/immunology , Atherosclerosis/pathology , Cells, Cultured , Disease Models, Animal , Endothelium, Vascular/diagnostic imaging , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Flow Cytometry , Fluorescent Dyes , Humans , Male , Mice , Microscopy, Confocal , Reproducibility of Results , Ultrasonography
11.
Invest Radiol ; 48(12): 843-50, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23857137

ABSTRACT

OBJECTIVES: The purposes of this study were the development and preclinical evaluation of clinically translatable E-selectin-specific ultrasound contrast agents based on a peptide ligand with the recognition sequence IELLQAR. MATERIALS AND METHODS: The E-selectin-specific peptide was synthesized through solid phase peptide synthesis and covalently attached to poly n-butylcyanoacrylate-stabilized microbubbles with an air core. Quantification of the microbubble surface coverage with peptides was performed through flow cytometry. Targeted adhesion of peptide-coated microbubbles was investigated in vitro using parallel plate flow chamber assays on tumor necrosis factor-α-stimulated human umbilical vein endothelial cells. In vivo imaging was performed in nude mice bearing human ovarian carcinoma xenografts (MLS), followed by ex vivo immunohistochemistry validation of E-selectin expression. RESULTS: Success of peptide synthesis was validated through preparative reverse phase high-pressure liquid chromatography and electronspray ionization-mass spectrometry. Results of the flow cytometry revealed approximately 4000 E-selectin-specific peptides/microbubble surface. Results of the in vitro experiments demonstrated the specificity of peptide-coated microbubbles to E-selectin (1.10 ± 0.48 vs 0.19 ± 0.09 bound microbubbles per cell, before and after competition respectively; P < 0.01). The in vivo imaging enabled specific assessment of E-selectin expression in MLS carcinoma xenografts (5.21 ± 3.41 vs 1.37 ± 0.67 contrast intensity before and after competition, respectively; P < 0.05). CONCLUSIONS: Clinically translatable microbubbles that were covalently coupled to the short E-selectin-specific peptide (IELLQAR) enabled specific imaging of the E-selectin expression in tumor vessels in vivo.


Subject(s)
E-Selectin/metabolism , Enbucrilate/chemistry , Molecular Imaging/methods , Ovarian Neoplasms/diagnostic imaging , Ovarian Neoplasms/metabolism , Peptides/pharmacokinetics , Ultrasonography/methods , Animals , Cell Line, Tumor , Drug Carriers/chemistry , Female , Mice , Mice, Nude , Microbubbles , Reproducibility of Results , Sensitivity and Specificity
12.
Article in English | MEDLINE | ID: mdl-23661125

ABSTRACT

The detection of microbubble contrast agents with ultrasound imaging techniques is the subject of ongoing research. Commonly, the nonlinear response of the agent is employed for detection. The performance of these techniques is, however, affected by nonlinear sound propagation. As an alternative, the change in echo response resulting from microbubble destruction can be employed to detect the agent. In this work, we propose a novel criterion for microbubble destruction detection that allows the rejection of tissue at a defined significance level even for highly echogenic structures in the presence of nonlinear propagation. Most clinical systems provide the hardware requirements for acquisitions consisting of multiple pulses transmitted at the same position, as used in Doppler imaging. Therefore, we develop a processing strategy that distinguishes contrast agent from other stationary or moving structures using these sequences. The proposed criterion is based on the variance of the phase shift of consecutive echoes in the sequence, which, in addition to tissue rejection, permits the distinction of motion from agent disruption. Phantom experiments are conducted to show the validity of the criterion and demonstrate the performance of the new method for contrast detection. Each detection series consists of 20 identical pulses at 9.5 MHz (4.7 MPa peak negative pressure) transmitted at a pulse repetition frequency of 5 kHz. The sequence is applied to phantoms under varied motion and flow conditions. As a first step toward molecular imaging, the technique is applied to microbubbles targeted to vascular endothelial growth factor receptor 2 (VEGFR2) in vitro. The results show a uniform rejection of the background signal while maintaining a contrast enhancement by more than 40 dB. The area under the receiver operating characteristics (ROC) curve is used as the performance metric for the separation of contrast agent and tissue signals, and values larger than 97% demonstrate that an excellent separation was achieved.


Subject(s)
Contrast Media/chemistry , Image Processing, Computer-Assisted/methods , Microbubbles , Signal Processing, Computer-Assisted , Ultrasonography/methods , Contrast Media/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Models, Biological , Phantoms, Imaging , ROC Curve , Reproducibility of Results , Signal-To-Noise Ratio , Vascular Endothelial Growth Factor Receptor-2/chemistry , Vascular Endothelial Growth Factor Receptor-2/metabolism
13.
J Control Release ; 163(1): 75-81, 2012 Oct 10.
Article in English | MEDLINE | ID: mdl-22580225

ABSTRACT

Microbubbles (MB) are routinely used contrast agents for functional and molecular ultrasound (US) imaging. In addition, they have been attracting more and more attention for drug delivery purposes, enabling e.g. US-mediated drug delivery across biological barriers and US-induced triggered drug release from the MB shell. The vast majority of efforts in this regard have thus far focused on phospholipid-based soft-shell MB, which are suboptimal for stably incorporating large amounts of drug molecules because of their relatively thin shell. Using poly(butyl cyanoacrylate) (PBCA)-based hard-shell MB, we show here that both hydrophilic (Rhodamine-B) and hydrophobic (Coumarin-6) model drugs can be efficiently and stably entrapped within the ~50 nm shell of PBCA MB. In addition, we demonstrate that model drug loading does not negatively affect the acoustic properties of the MB, and that functionalizing the surface of fluorophore-loaded MB with anti-VEGFR2 antibodies enables image-guided and targeted model drug delivery to tumor blood vessels. Finally, we show both in vitro and in vivo that disintegrating VEGFR2-targeted MB with high-mechanical index US pulses leads to high levels of model drug release. Consequently, these findings indicate that polymer-based MB are highly suitable systems for image-guided, targeted and triggered drug delivery to tumors and tumor blood vessels.


Subject(s)
Cyanoacrylates/chemistry , Drug Delivery Systems , Microbubbles , Neoplasms/metabolism , Acoustics , Animals , Cell Line, Tumor , Coumarins/administration & dosage , Coumarins/chemistry , Enbucrilate , Fluorescent Dyes/administration & dosage , Fluorescent Dyes/chemistry , Mice , Microscopy, Fluorescence , Rhodamines/administration & dosage , Rhodamines/chemistry , Thiazoles/administration & dosage , Thiazoles/chemistry , Vascular Endothelial Growth Factor Receptor-2/metabolism
14.
J Nucl Med ; 53(3): 345-8, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22393225

ABSTRACT

Ultrasound imaging is clinically established for routine screening examinations of breast, abdomen, neck, and other soft tissues, as well as for therapy monitoring. Microbubbles as vascular contrast agents improve the detection and characterization of cancerous lesions, inflammatory processes, and cardiovascular pathologies. Taking advantage of the excellent sensitivity and specificity of ultrasound for microbubble detection, molecular imaging can be realized by binding antibodies, peptides, and other targeting moieties to microbubble surfaces. Molecular microbubbles directed against various targets such as vascular endothelial growth factor receptor-2, vascular cell adhesion molecule 1, intercellular adhesion molecule 1, selectins, and integrins were developed and were shown in preclinical studies to be able to selectively bind to tumor blood vessels and atherosclerotic plaques. Currently, the first microbubble formulations targeted to angiogenic vessels in prostate cancers are being evaluated clinically. However, microbubbles can be used for more than diagnosis: disintegrating microbubbles emit acoustic forces that are strong enough to induce thrombolysis, and they can also be used for facilitating drug and gene delivery across biologic barriers. This review on the use of microbubbles for ultrasound-based molecular imaging, therapy, and theranostics addresses innovative concepts and identifies areas in which clinical translation is foreseeable in the near future.


Subject(s)
Contrast Media , Microbubbles , Molecular Diagnostic Techniques , Thrombolytic Therapy/methods , Ultrasonics , Animals , Clinical Trials as Topic , Drug Delivery Systems , Genetic Therapy/methods , Humans , Vascular Endothelial Growth Factor Receptor-2/metabolism
15.
Curr Pharm Des ; 18(15): 2184-99, 2012.
Article in English | MEDLINE | ID: mdl-22352772

ABSTRACT

Ultrasound is one of the workhorses in clinical cancer diagnosis. In particular, it is routinely used to characterize lesions in liver, urogenital tract, head and neck and soft tissues. During the last years image quality steadily improved, which, among others, can be attributed to the development of harmonic image analysis. Microbubbles were introduced as intravascular contrast agents and can be detected with superb sensitivity and specificity using contrast specific imaging modes. By aid of these unspecific contrast agents tissues can be characterised regarding their vascularity. Antibodies, peptides and other targeting moieties were bound to microbubbles to target sites of angiogenesis and inflammation intending to get more disease-specific information. Indeed, many preclinical studies proved the high potential of targeted ultrasound imaging to better characterize tumors and to more sensitively monitor therapy response. Recently, first targeted microbubbles had been developed that meet the pharmacological demands of a clinical contrast agent. This review articles gives an overview on the history and current status of targeted ultrasound imaging of cancer. Different imaging concepts and contrast agent designs are introduced ranging from the use of experimental nanodroplets to agents undergoing clinical evaluation. Although it is clear that targeted ultrasound imaging works reliably, its broad acceptance is hindered by the user dependency of ultrasound imaging in general. Automated 3D-scanning techniques-like being used for breast diagnosis - and novel 3D transducers will help to make this fascinating method clinical reality.


Subject(s)
Contrast Media , Microbubbles , Neoplasms/diagnostic imaging , Animals , Humans , Imaging, Three-Dimensional/methods , Nanoparticles , Particle Size , Sensitivity and Specificity , Ultrasonography
16.
Drug Deliv Transl Res ; 2(1): 56-64, 2012 Feb.
Article in English | MEDLINE | ID: mdl-25786599

ABSTRACT

Microbubbles (MB) are routinely used as contrast agents for functional and molecular ultrasound (US) imaging. For molecular US imaging, MB are functionalized with antibodies or peptides, in order to visualize receptor expression by angiogenic or inflamed endothelium. In general, initial in vitro binding studies with targeted MB are performed using phase contrast microscopy. Difficulties in the identification of MB in standard phase contrast microscopy, however, generally result in high variability, high observer dependency, and low reproducibility. To overcome these shortcomings, we here describe a simple post-loading strategy for labeling polymer-based MB with fluorophores, and we show that the use of rhodamine-loaded MB in combination with fluorescence microscopy substantially reduces the variability and the observer dependency of in vitro binding studies. In addition, we demonstrate that rhodamine-loaded MB can also be used for in vivo and ex vivo experimental setups, e.g., for analyzing MB binding to inflamed carotids using two-photon laser scanning microscopy, and for validating the binding of VEGFR2-targeted MB to tumor endothelium. These findings demonstrate that fluorescently labeled MB substantially facilitate translational molecular US studies, and they suggest that a similar synthetic strategy can be exploited for preparing drug-loaded MB, to enable image-guided, targeted, and triggered drug delivery to tumors and to sites of inflammation.

17.
Eur J Radiol ; 81(10): 2710-6, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22093958

ABSTRACT

PURPOSE: To compare non-enhanced and contrast-enhanced high-frequency 3D Doppler ultrasound with contrast-enhanced 2D and 3D B-mode imaging for assessing tumor vascularity during antiangiogenic treatment using soft-shell and hard-shell microbubbles. MATERIALS AND METHODS: Antiangiogenic therapy effects (SU11248) on vascularity of subcutaneous epidermoid-carcinoma xenografts (A431) in female CD1 nude mice were investigated longitudinally using non-enhanced and contrast-enhanced 3D Doppler at 25 MHz. Additionally, contrast-enhanced 2D and 3D B-mode scans were performed by injecting hard-shell (poly-butyl-cyanoacrylate-based) and soft-shell (phospholipid-based) microbubbles. Suitability of both contrast agents for high frequency imaging and the sensitivity of the different ultrasound methods to assess early antiangiogenic therapy effects were investigated. Ultrasound data were validated by immunohistology. RESULTS: Hard-shell microbubbles induced higher signal intensity changes in tumors than soft-shell microbubbles in 2D B-mode measurements (424 ± 7 vs. 169 ± 8 A.U.; p<0.01). In 3D measurements, signals of soft-shell microbubbles were hardly above the background (5.48 ± 4.57 vs. 3.86 ± 2.92 A.U.), while signals from hard-shell microbubbles were sufficiently high (30.5 ± 8.06 A.U). Using hard-shell microbubbles 2D and 3D B-mode imaging depicted a significant decrease in tumor vascularity during antiangiogenic therapy from day 1 on. Using soft-shell microbubbles significant therapy effects were observed at day 4 after therapy in 2D B-mode imaging but could not be detected in the 3D mode. With non-enhanced and contrast-enhanced Doppler imaging significant differences between treated and untreated tumors were found from day 2 on. CONCLUSION: Hard-shell microbubble-enhanced 2D and 3D B-mode ultrasound achieved highest sensitivity for assessing therapy effects on tumor vascularisation and were superior to B-mode ultrasound with soft-shell microbubbles and to Doppler imaging.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Carcinoma, Squamous Cell/diagnostic imaging , Carcinoma, Squamous Cell/drug therapy , Imaging, Three-Dimensional/methods , Microbubbles , Neovascularization, Pathologic/diagnostic imaging , Animals , Carcinoma, Squamous Cell/complications , Cell Line, Tumor , Contrast Media , Female , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/etiology , Reproducibility of Results , Sensitivity and Specificity , Treatment Outcome , Ultrasonography/methods
18.
Ultrasound Med Biol ; 37(10): 1622-34, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21924206

ABSTRACT

We aimed to develop and characterize poly n-butylcyanoacrylate (PBCA) microbubbles (MBs) with a narrow size distribution. MBs were synthesized by established emulsion polymerization techniques, size-isolated by centrifugation and functionalized for molecular imaging by coating their surface with streptavidin. The physical and acoustic properties of the parent solution, different-size isolated populations and functionalized MBs were measured and compared. As expected from negative zeta potentials at pH 7, cryo scanning electron microscopy showed no aggregates. In phantoms MBs were destructible at high mechanical indices and showed a frequency-dependent attenuation and backscattering. The MBs were stable in solution for more than 14 weeks and could be lyophilized without major damage. However, for injection, small needle diameters and high injection rates are shown to be critical because both lead to MB destruction. In summary, when being handled correctly, size-isolated PBCA MBs are promising candidates for preclinical functional and molecular ultrasound imaging.


Subject(s)
Contrast Media/chemical synthesis , Enbucrilate/chemistry , Ultrasonography , Centrifugation , Coated Materials, Biocompatible/chemistry , Drug Stability , Flow Cytometry , Freeze Drying , Microbubbles , Microscopy, Electron, Scanning , Streptavidin/chemistry
19.
Biomaterials ; 32(26): 6155-63, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21632103

ABSTRACT

Magnetic resonance (MR) and ultrasound (US) imaging are widely used diagnostic modalities for various experimental and clinical applications. In this study, iron oxide nanoparticle-embedded polymeric microbubbles were designed as multi-modal contrast agents for hybrid MR-US imaging. These magnetic nano-in-micro imaging probes were prepared via a one-pot emulsion polymerization to form poly(butyl cyanoacrylate) microbubbles, along with the oil-in-water (O/W) encapsulation of iron oxide nanoparticles in the bubble shell. The nano-in-micro embedding strategy was validated using NMR and electron microscopy. These hybrid imaging agents exhibited strong contrast in US and an increased transversal relaxation rate in MR. Moreover, a significant increase in longitudinal and transversal relaxivities was observed after US-induced bubble destruction, which demonstrated triggerable MR imaging properties. Proof-of-principle in vivo experiments confirmed that these nanoparticle-embedded microbubble composites are suitable contrast agents for both MR and US imaging. In summary, these magnetic nano-in-micro hybrid materials are highly interesting systems for bimodal MR-US imaging, and their enhanced relaxivities upon US-induced destruction recommend them as potential vehicles for MR-guided US-mediated drug and gene delivery.


Subject(s)
Contrast Media/chemistry , Ferric Compounds/chemistry , Magnetic Resonance Imaging/methods , Microbubbles , Nanoparticles/chemistry , Polymers/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...