Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Genes Brain Behav ; 16(7): 686-698, 2017 09.
Article in English | MEDLINE | ID: mdl-28387990

ABSTRACT

Genome-wide association studies in humans have suggested that variants of the cadherin-13 (CDH13) gene are associated with substance use disorder, subjective response to amphetamine, and attention deficit hyperactivity disorder. To examine the role of the Cdh13 and its peptide ligand adiponectin (AdipoQ) in addiction-related behaviors, we assessed Cdh13 knockout (KO) rats and AdipoQ KO mice using intravenous cocaine self-administration and conditioned place preference (CPP) paradigms. During intravenous cocaine self-administration, male Cdh13 heterozygous (+/-) and KO (-/-) rats showed increased cue-induced reinstatement compared with wild-type (WT) rats when presented with a cocaine-paired stimulus, whereas female Cdh13 rats showed no differences across genotype. Cdh13 -/- rats showed higher responding for a saccharin reinforcer and learned the choice reaction time (RT) task more slowly than WTs. However, we found no differences between Cdh13 -/- and +/+ rats in responding for sensory reinforcement, number of premature responses in the RT task, tendency to approach a Pavlovian food cue, CPP and locomotor activation to cocaine (10 or 20 mg/kg). In AdipoQ -/- mice, there was a significant increase in CPP to methamphetamine (1 mg/kg) but not to a range of d-amphetamine doses (0.5, 1, 2 and 4 mg/kg). Taken together, these data suggest that Cdh13 and AdipoQ regulate sensitivity to psychomotor stimulants and palatable rewards without producing major changes in other behaviors. In humans, these two genes may regulate sensitivity to natural and drug rewards, thus influencing susceptibility to the conditioned drug effects and relapse.


Subject(s)
Adiponectin/genetics , Amphetamine-Related Disorders/genetics , Cadherins/genetics , Cocaine-Related Disorders/genetics , Conditioning, Classical , Conditioning, Operant , Reward , Adiponectin/metabolism , Animals , Cadherins/metabolism , Choice Behavior , Cues , Female , Male , Rats , Rats, Inbred Dahl , Reaction Time
2.
J Control Release ; 256: 1-8, 2017 06 28.
Article in English | MEDLINE | ID: mdl-28412225

ABSTRACT

High Intensity Focused Ultrasound (HIFU) is an emerging noninvasive, nonionizing physical energy based modality to ablate solid tumors with high power, or increase local permeability in tissues/tumors in pulsed mode with relatively low power. Compared with traditional ablative HIFU, nondestructive pulsed HIFU (pHIFU) is present in the majority of novel applications recently developed for enhancing the delivery of drugs and genes. Previous studies have demonstrated the capability of pHIFU to change tissue local permeability for enhanced drug delivery in both mouse tumors and mouse muscle. Further study based on bulk tissues in large animals and clinical HIFU system revealed correlation between therapeutic effect and thermal parameters, which was absent in the previous mouse studies. In this study, we further investigated the relation between the therapeutic effect of pHIFU and thermal parameters in bulky normal muscle tissues based on a rabbit model and a preclinical HIFU system. Correlation between therapeutic effect and thermal parameters was confirmed in our study on the same bulk tissues although different HIFU systems were used. Following the study in bulky normal muscle tissues, we further created bulky tumor model with VX2 tumors implanted on both hind limbs of rabbits and investigated the feasibility to enhance tumor permeability in bulky VX2 tumors in a rabbit model using pHIFU technique. A radiolabeled peptidomimetic integrin antagonist, 111In-DOTA-IA, was used following pHIFU treatment in our study to target VX2 tumor and serve as the radiotracer for follow-up single-photon emission computed tomography (SPECT) scanning. The results have shown significantly elevated uptake of 111In-DOTA-IA in the area of VX2 tumors pretreated by pHIFU compared with the control VX2 tumors not being pretreated by pHIFU, and statistical analysis revealed averaged 34.5% enhancement 24h after systematic delivery of 111In-DOTA-IA in VX2 tumors pretreated by pHIFU compared with the control VX2 tumors.


Subject(s)
Coordination Complexes/administration & dosage , Drug Delivery Systems , Heterocyclic Compounds, 1-Ring/administration & dosage , High-Intensity Focused Ultrasound Ablation , Indium Radioisotopes/administration & dosage , Muscle Neoplasms , Animals , Buttocks/diagnostic imaging , Coordination Complexes/pharmacokinetics , Coordination Complexes/therapeutic use , Female , Heterocyclic Compounds, 1-Ring/pharmacokinetics , Heterocyclic Compounds, 1-Ring/therapeutic use , Indium Radioisotopes/pharmacokinetics , Indium Radioisotopes/therapeutic use , Magnetic Resonance Imaging , Muscle Neoplasms/diagnostic imaging , Muscle Neoplasms/metabolism , Muscle Neoplasms/therapy , Permeability , Rabbits , Tomography, Emission-Computed, Single-Photon
3.
J Control Release ; 217: 113-20, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26334482

ABSTRACT

The blood-brain barrier (BBB), comprised of brain endothelial cells with tight junctions (TJ) between them, regulates the extravasation of molecules and cells into and out of the central nervous system (CNS). Overcoming the difficulty of delivering therapeutic agents to specific regions of the brain presents a major challenge to treatment of a broad range of brain disorders. Current strategies for BBB opening are invasive, not specific, and lack precise control over the site and timing of BBB opening, which may limit their clinical translation. In the present report, we describe a novel approach based on a combination of stem cell delivery, heat-inducible gene expression and mild heating with high-intensity focused ultrasound (HIFU) under MRI guidance to remotely permeabilize BBB. The permeabilization of the BBB will be controlled with, and limited to where selected pro-inflammatory factors will be secreted secondary to HIFU activation, which is in the vicinity of the engineered stem cells and consequently both the primary and secondary disease foci. This therapeutic platform thus represents a non-invasive way for BBB opening with unprecedented spatiotemporal precision, and if properly and specifically modified, can be clinically translated to facilitate delivery of different diagnostic and therapeutic agents which can have great impact in treatment of various disease processes in the central nervous system.


Subject(s)
Blood-Brain Barrier/metabolism , Stem Cells , Animals , Cells, Cultured , Gene Expression , Genetic Vectors , Green Fluorescent Proteins/genetics , HEK293 Cells , HSP70 Heat-Shock Proteins/genetics , Hot Temperature , Humans , Lentivirus/genetics , Luciferases/metabolism , Magnetic Resonance Imaging , Male , Mice , Permeability , Rats, Nude , Transgenes , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Ultrasonic Therapy
4.
Mol Pharm ; 10(10): 3603-11, 2013 Oct 07.
Article in English | MEDLINE | ID: mdl-23961901

ABSTRACT

Multivalency is a powerful strategy for achieving high-affinity molecular binding of compounds to increase their therapeutic potency or imaging potential. In our study, multivalent non-peptide integrin αvß3 antagonists (IA) were designed for antitumor therapy. Docking and molecular dynamics were employed to explore the binding modes of IA monomer, dimer, and trimer. In silico, one IA unit binds tightly in the active site with similar pose to native ligand RGD and other parts of dimer and trimer contribute extra binding affinities by interacting with residues in vicinity of the original site. In vitro studies demonstrated that increasing valency results in increasing antiproliferative and antiorganizational effects against endothelial cells (HUVECs), and a much weaker effect on melanoma B16F10 cells. The antitumor efficacies of the IA multivalent compounds were evaluated in subcutaneous B16F10 melanoma tumor-bearing mice. At 30 mg/kg dose, the mean masses of tumors harvested 18 days after inoculation were significantly reduced (p<10(-7)) by 36±9%, 49±8%, and 71±7% for the IA monomer, dimer, and trimer groups, relative to control. The importance of multivalency was demonstrated to be highly significant beyond the additive effect of the extra pharmacological sites (p=0.00011). These results suggest that the major target of these anti-αvß3 compounds is the neovasculature rather than the cancer cells, and the success of a multivalent strategy depends on the details of the components and linker. This is the first integrin αvß3 multivalent ligand showing clear enhancement in antitumor effectiveness.


Subject(s)
Antineoplastic Agents/pharmacology , Integrin alphaVbeta3/antagonists & inhibitors , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Computational Biology , Enzyme-Linked Immunosorbent Assay , Humans , Molecular Dynamics Simulation , Protein Structure, Secondary
5.
Mol Biosyst ; 9(3): 398-406, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23287963

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a devastating and chronic neurodegenerative disease without any known cure. In the brain and spinal cord of both patients and animal models with ALS, neuroinflammation is a prominent pathological hallmark which is characterized by infiltrating T cells at sites of motor neuron injury. Their presence in mutant Cu(2+)/Zn(2+) superoxide dismutase (mSOD1) induced ALS plays an important role in shifting the response of microglia from neuroprotective to neurotoxic. In order to better understand how these cells and their communication network collectively modulate the disease progression, we have established a mathematical model integrating diverse cells and cytokines. According to the experimental data sets, we first refined this model by identifying a link between TGFß and M1 microglia which can produce an optimized model to fit data sets better. Then based on this model, parameters were estimated using genetic algorithm. Sensitivity analysis of these parameters identified several factors such as the release rate of IFNγ by T helper 1 (Th1) cells, which may be related to the heterogeneity between the patients with different survival times. Furthermore, the tests on T cell based therapeutic strategies indicated that elimination of Th1 cells is the most effective approach extending survival time. This confirmed the dominant role of Th1 cells in leading the rapid disorder in the later stage of ALS. For the therapies targeting cytokines, injection of IL6 can essentially augment the neuroprotective response and extend the life effectively by elevating the level of IL4, a neuroprotective cytokine, while directly injected IL4 will decay rapidly in the ALS microenvironment and cannot provide a persistent protective effect. On the other hand, in spite of the attractive effect of direct elimination of mSOD1 or self-antigen, it is difficult to implement in CNS. As an alternative, elimination of IFNγ can be chosen as another effective therapy. In the future, if we combine the side effects of different therapies, this model can be used to optimize the therapeutic strategies so that they can effectively improve survival rates and quality of life for patients with ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Cell Communication , Computer Simulation , Models, Biological , Algorithms , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/enzymology , Amyotrophic Lateral Sclerosis/immunology , Animals , Cytokines/pharmacology , Cytokines/physiology , Humans , Metabolic Networks and Pathways , Mice , Microglia/drug effects , Microglia/pathology , Motor Neurons/drug effects , Motor Neurons/pathology , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , T-Lymphocytes, Regulatory/immunology , Th2 Cells/immunology
6.
AJR Am J Roentgenol ; 197(2): 318-24, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21785076

ABSTRACT

OBJECTIVE: A theranostic system integrates some form of diagnostic testing to determine the presence of a molecular target for which a specific drug is intended. Molecular imaging serves this diagnostic function and provides powerful means for noninvasively detecting disease. We briefly review the paradigms rooted in nuclear medicine and highlight recent advances in this field. We also explore how nanometer-sized complexes, called nanomedicines, present an excellent theranostic platform applicable to both drug discovery and clinical use. CONCLUSION: For imagers, molecular theranostics represents a powerful emerging platform that intimately couples targeted therapeatic entities with noninvasive imaging that yields information on the presence of defined molecular targets before, during, and after cognate therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Molecular Imaging/methods , Nanomedicine/methods , Neoplasms/diagnosis , Neoplasms/drug therapy , Nuclear Medicine/methods , Contrast Media , Humans , Molecular Probe Techniques , Radiopharmaceuticals
7.
Curr Radiopharm ; 3(1): 1-8, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-20556233

ABSTRACT

OBJECTIVES: The aim of this research was to synthesize radiolabeled peptidomimetic integrin alpha(v)beta(3) antagonist with (99m)Tc for rapid targeting of integrin alpha(v)beta(3) receptors in tumor to produce a high tumor to background ratio. METHODS: The amino terminus of 4-[2-(3,4,5,6-tetra-hydropyrimidin-2-ylamino)-ethyloxy]benzoyl-2-(S)-[N-(3-amino-neopenta-1-carbamyl)]-aminoethylsulfonyl-amino-beta-alanine hydrochloride (IAC) was conjugated with N-hydroxysuccinimide ester of HYNIC and labeled with (99m)Tc using tricine with either 1,5-pyridinedicarboxylic acid (PDA) or ethylenediamine-N,N'-diacetic acid (EDDA) as the co-ligand. The products, (99m)Tc EDDA(2)/HYNIC-IAC (P1) and (99m)Tc PDA (tricin)/HYNIC-IAC (P2) were subjected to in vitro serum stability, receptor-binding, biodistribution and imaging studies. RESULTS: P1 and P2 were synthesized with an overall yield of >80%. P1 was slightly more stable than P2 when incubated in serum at 37 degrees C for 18 hrs (84 vs 77% intact). The In vitro receptor-binding of P1 was higher than that of P2 (78.02 +/- 13.48 vs 51.05 +/- 14.05%) when incubated with alpha(v)beta(3) at a molar excess (0.8 muM). This receptor binding was completely blocked by a molar excess of an unlabeled peptidomimetic antagonist. Their differences shown in serum stability and the receptor-binding appeared to be related to their biological behaviors in tumor uptake and retention; the 1 h tumor uptakes of P1 and P2 were 3.17+/-0.52 and 2.13+/-0.17 % ID/g, respectively. P1 was retained in the tumor longer than P2. P1 was excreted primarily through the renal system whereas P2 complex was excreted equally via both renal and hepatobiliary systems. Thus, P1 was retained in the whole-body with 27.25 +/- 3.67% ID at 4 h whereas 54.04 +/- 3.57% ID of P2 remained in the whole-body at 4 h. This higher whole-body retention of P2 appeared to be resulted from a higher amount of radioactivity retained in liver and intestine. These findings were supported by imaging studies showing higher tumor-to-abdominal contrast for P1 than for P2 at 3 h postinjection. CONCLUSIONS: P1 showed good tumor targeting properties with a rapid tumor uptake, prolonged tumor retention and fast whole-body clearance kinetics. These findings warrant further investigation of the HYNIC method of (99m)Tc labeling of other peptidomimetic antagonists using EDDA as a coligand.

8.
Bioconjug Chem ; 21(2): 270-8, 2010 Feb 17.
Article in English | MEDLINE | ID: mdl-20102181

ABSTRACT

Computer modeling approaches to identify new inhibitors are essentially a very sophisticated and efficient way to design drugs. In this study, a bivalent nonpeptide intergrin alpha(v)beta(3) antagonist (bivalent IA) has been synthesized on the basis of an in silico rational design approach. A near-infrared (NIR) fluorescent imaging probe has been developed from this bivalent compound. In vitro binding assays have shown that the bivalent IA (IC(50) = 0.40 +/- 0.11 nM) exhibited improved integrin alpha(v)beta(3) affinity in comparison with the monovalent IA (IC(50) = 22.33 +/- 4.51 nM), resulting in an over 50-fold improvement in receptor affinity. NIR imaging probe, bivalent-IA-Cy5.5 conjugate, also demonstrated significantly increased binding affinity (IC(50) = 0.13 +/- 0.02 nM). Fluorescence microscopy studies showed integrin-mediated endocytosis of bivalent-IA-Cy5.5 in U87 cells which was effectively blocked by nonfluorescent bivalent IA. We also demonstrated tumor accumulation of this NIR imaging probe in U87 mouse xenografts.


Subject(s)
Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/metabolism , Infrared Rays , Integrin alphaVbeta3/antagonists & inhibitors , Molecular Imaging/methods , Neoplasms/metabolism , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , Computer Simulation , Early Detection of Cancer , Female , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Integrin alphaVbeta3/chemistry , Integrin alphaVbeta3/metabolism , Mice , Microscopy, Fluorescence , Models, Molecular , Neoplasm Metastasis , Neoplasms/diagnosis , Neoplasms/genetics , Neoplasms/pathology , Protein Conformation , Substrate Specificity
9.
J Biomed Opt ; 14(3): 034032, 2009.
Article in English | MEDLINE | ID: mdl-19566325

ABSTRACT

Dye-labeled protein microspheres, submicron in size and capable of producing thermoelastically generated ultrasound in response to laser stimulation, are presented as contrast agents for photoacoustic imaging. Incident laser energy absorbed by fluorescein isothiocyanate (FITC)-labeled elastin submicrospheres results in thermoelastically generated sound production. Plotted A-line graphs reveal a distinctive morphology and a greater than two orders of magnitude increase in signal amplitude subsequent to converting FITC elastin into submicrospheres (despite a four orders of magnitude decrease in concentration). Evidence of nonlinearity and enhancement of ultrasound backscatter indicate a potential use in contrast-enhanced harmonic imaging. Photoacoustic and ultrasound imaging of FITC-elastin submicrospheres in a water-filled phantom vessel shows enhanced contrast at low concentration and clear delineation of the phantom vessel wall.


Subject(s)
Coloring Agents/chemistry , Contrast Media/chemistry , Optics and Photonics/methods , Proteins/chemistry , Ultrasonography/methods , Equipment Design , Fluorescein-5-isothiocyanate/chemistry , Microspheres , Particle Size , Phantoms, Imaging , Spectrometry, Fluorescence/methods , Water/chemistry
10.
AJR Am J Roentgenol ; 193(2): 287-94, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19620423

ABSTRACT

OBJECTIVE: Molecular imaging has emerged as a powerful technology that has already changed the practice of modern medicine. During this same period, the monumental genome project has sequenced man's entire genetic content. Now the postgenomic aim is to understand the dynamic interactions of the encoded components and their regulatory mechanisms. CONCLUSION: Molecular imaging is well positioned to play a major role in this massive effort as we move toward a comprehensive paradigm for assessing health and disease.


Subject(s)
Diagnostic Techniques and Procedures , Molecular Probe Techniques , Systems Biology , Computational Biology/methods , Human Genome Project , Humans , Knowledge Bases , Molecular Sequence Data
11.
Eur J Radiol ; 70(2): 201-4, 2009 May.
Article in English | MEDLINE | ID: mdl-19261418

ABSTRACT

The term "Molecular Imaging" has hit the consciousness of radiologists only in the past decade although many of the concepts that molecular imaging encompasses has been practiced in biomedical imaging, especially in nuclear medicine, for many decades. Many new imaging techniques have allowed us to interrogate biologic events at the cellular and molecular level in vivo in four dimensions but the challenge now is to translate these techniques into clinical practice in a way that will enable us to revolutionize healthcare delivery. The purpose of this article is to introduce the term "Systems Diagnostics" and examine how radiologists can become translators of disparate sources of information into medical decisions and therapeutic actions.


Subject(s)
Algorithms , Diagnostic Imaging/trends , Gene Expression Profiling/trends , Molecular Probe Techniques/trends
12.
Int J Hyperthermia ; 24(6): 506-20, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18608574

ABSTRACT

This paper reviews the enhanced delivery of genes, drugs and therapeutics using ultrasound. It begins with a general overview of the field and the various techniques associated with it, including sonophoresis, hyperthermia (with ultrasound), sonoporation, and microbubble assisted transvascular and targeted delivery. Particular attention is then paid to pulsed high intensity focused ultrasound drug delivery without the use of ultrasound contrast agents. Feasibility and mechanistic studies of this technique are described in some detail. Conclusions are then drawn regarding possible mechanisms of this treatment, and to contrast with the better known treatments relying on injection of ultrasound contrast agents.


Subject(s)
Drug Delivery Systems/methods , Ultrasonics , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Contrast Media/chemistry , Gene Transfer Techniques , Humans , Hyperthermia, Induced/methods , Microbubbles , Neoplasms/pathology , Neoplasms/therapy , Thrombolytic Therapy/methods , Tumor Necrosis Factor-alpha/metabolism
13.
Radiology ; 248(2): 485-91, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18574138

ABSTRACT

PURPOSE: To investigate whether combining pulsed high-intensity focused ultrasound (HIFU) with the chemotherapeutic drug bortezomib could improve antitumor activity against murine squamous cell carcinoma (SCC) tumors. MATERIALS AND METHODS: All experiments were conducted with animal care and use committee approval. Murine SCC cells were implanted subcutaneously in C3H mice. When tumors reached 100 mm(3), mice were randomized to one of three groups for twice weekly intraperitoneal injections of 1.5 mg of bortezomib per kilogram of body weight, a proteasome inhibitor (n = 10); 1.0 mg/kg bortezomib (n = 11); or a control vehicle (n = 12). Within each group, half of the mice received pulsed HIFU exposure to their tumors immediately prior to each injection. The time for tumors to reach 650 mm(3) was compared among groups. Additional tumors were stained with terminal deoxynucledotidyl transferase-mediated dUTP nick end labeling and CD31 to assess apoptotic index and blood vessel density, respectively. RESULTS: Tumors in the control group, pulsed HIFU and control group, and 1.0 mg/kg of bortezomib alone group reached the size end point in 5.2 days +/- 0.8 (standard deviation), 5.3 days +/- 0.8, and 5.6 days +/- 1.1, respectively. However, pulsed HIFU and 1.0 mg/kg bortezomib increased the time to end point to 9.8 days +/- 2.9 (P < .02), not significantly different from the 8.8 days +/- 2.1 in tumors treated with 1.5 mg/kg bortezomib alone (P > .05). Combination therapy was also associated with a significantly higher apoptotic index (P < .05). CONCLUSION: Treatment of tumors with pulsed HIFU lowered the threshold level for efficacy of bortezomib, resulting in significant tumor cytotoxicity and growth inhibition at lower dose levels.


Subject(s)
Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Carcinoma, Squamous Cell/therapy , Pyrazines/pharmacology , Ultrasonic Therapy , Animals , Apoptosis , Bortezomib , Carcinoma, Squamous Cell/drug therapy , Combined Modality Therapy , Mice , Random Allocation , Transplantation, Heterologous , Tumor Cells, Cultured
14.
J Nucl Med ; 49(2): 295-302, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18199622

ABSTRACT

UNLABELLED: The aim of this study was to determine if pulsed high-intensity focused ultrasound (HIFU) exposures could enhance tumor uptake of (111)In-MX-B3, a murine IgG1kappa monoclonal antibody directed against the Le(y) antigen. METHODS: MX-B3 was labeled with (111)In, purified, and confirmed for its binding to the antigen-positive A431 cell line. Groups of nude mice were inoculated subcutaneously with A431 tumor cells on both hind flanks. A tumor on one flank was treated with pulsed-HIFU; the other tumor was used as an untreated control. Within 10 min after the HIFU exposure, the mice received intravenous (111)In-MX-B3 for imaging and biodistribution studies. Mice were euthanized at 1, 24, 48, and 120 h after injection for biodistribution studies. RESULTS: The HIFU exposure shortened the peak tumor uptake time (24 vs. 48 h for the control) and increased the peak tumor uptake value (38 vs. 25 %ID/g [percentage injected dose per gram] for the control). The HIFU effect on enhancing tumor uptake was greater at earlier times up to 24 h, but the effect was gradually diminished thereafter. The HIFU effect on enhancing tumor uptake was substantiated by nuclear imaging studies. HIFU also increased the uptake of the antibody in surrounding tissues, but the net increase was marginal compared with the increase in tumor uptake. CONCLUSION: This study demonstrates that pulsed-HIFU significantly enhances the delivery of (111)In-MX-B3 in human epidermoid tumors xenografted in nude mice. The results of this pilot study warrant further evaluation of other treatment regimens, such as repeated HIFU exposures for greater delivery enhancement of antibodies labeled with cytotoxic radioisotopes or pulsed-HIFU exposure in addition to a combined therapy of (90)Y-B3 and taxol to enhance the synergistic effect.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Phonophoresis/methods , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Carcinoma, Squamous Cell/diagnostic imaging , Carcinoma, Squamous Cell/therapy , Humans , Isotope Labeling , Metabolic Clearance Rate/radiation effects , Mice , Mice, Nude , Organ Specificity/radiation effects , Radioimmunotherapy/methods , Radionuclide Imaging , Tissue Distribution/radiation effects
15.
Int J Nanomedicine ; 2(3): 479-85, 2007.
Article in English | MEDLINE | ID: mdl-18019845

ABSTRACT

Angiogenesis is an important process in cancer growth and metastasis. During the tumor angiogenic process, endothelial cells express various cell surface receptors which can be utilized for molecular imaging and targeted drug delivery. One such protein receptor of interest is the integrin alphav beta3. Our group is involved in the development of molecular imaging probes and drug delivery systems targeting alphav beta3. Based on extensive lead optimization study with the integrin antagonist compounds, we have developed a new generation of integrin alphav beta3 compound (IA) which has superior binding affinity to alphav beta3. Utilizing this IA as a targeting agent, we have developed a novel integrin-targeted nanoparticle (ITNP) system for targ alphav beta3 was observed. These ITNPs also were rapidly taken up by cells that express alphav beta3. The ITNPs accumulated in the angiogenic vessels, after systemic administration in a murine squamous cell carcinoma model. This novel intergrin targeted ITNP platform will likely have an application in targeted delivery of drugs and genes in vivo and can also be used for molecular imaging.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Endothelial Cells/metabolism , Integrin alphaVbeta3/antagonists & inhibitors , Nanoparticles/chemistry , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Angiogenesis Inhibitors/pharmacokinetics , Animals , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Drug Delivery Systems/methods , Endothelial Cells/drug effects , Humans , Mice , Mice, Inbred C3H
16.
Nucl Med Biol ; 34(4): 363-70, 2007 May.
Article in English | MEDLINE | ID: mdl-17499725

ABSTRACT

OBJECTIVES: The aim of this research was to synthesize radiolabeled peptidomimetic integrin alpha(v)beta(3) antagonists that selectively target integrin alpha(v)beta(3) receptor and clear rapidly from the whole body. METHODS: Integrin alpha(v)beta(3) antagonists, 4-[2-(3,4,5,6-tetrahydropyrimidine-2-ylamino)ethyloxy]benzoyl-2-(S)-aminoethylsulfonyl-amino-beta-alanine (IA) and 4-[2-(3,4,5,6-tetrahydro-pyrimidin-2-ylamino)-ethyloxy]benzoyl-2-(S)-[N-(3-amino-neopenta-1-carbamyl)]-aminoethylsulfonylamino-beta-alanine hydrochloride (IAC), a hydrophobic carbamate derivative of IA, were conjugated with 2-p-isothiocyanatobenzyl-DOTA at the amino terminus and labeled with (111)In. The (111)In labeled IA and IAC were subjected to in vitro receptor binding, biodistribution and imaging studies using nude mice bearing the receptor-positive M21 human melanoma xenografts. RESULTS: The (111)In-labeled IA (40%) and -IAC (72%) specifically bound in vitro to alpha(v)beta(3) (0.8 microM) at a molar excess. This receptor binding was completely blocked by a molar excess of cold IA to alpha(v)beta(3). The higher receptor-binding affinity of the (111)In-labeled IAC was reflected in higher tumor uptake and retention: 5.6+/-1.4 and 4.5+/-0.7 %ID/g vs. 3.8+/-0.9 and 2.0+/-0.3 %ID/g for the (111)In-labeled IA at 0.33 and 2 h. The tumor uptakes were inhibited by the co-injection of 200 microg of IA, indicating that the uptake was receptor mediated. These antagonists were excreted primarily via the renal system. The (111)In activity retained in the whole body was quite comparable between the (111)In-labeled IA (24% ID) and the (111)In-labeled IAC (33% ID) at 2 h. The higher peak tumor uptake and longer retention resulted in higher tumor-to-background ratios for the (111)In-labeled IAC at 2 h with 9.7, 2.3, 0.8, 1.9, 7.1, 2.2, 0.9, 3.7 and 9.9 for blood, liver, kidney, lung, heart, stomach, intestine, bone and muscle, respectively. The imaging studies with the (111)In-labeled IAC also clearly visualized the receptor-positive tumor at 4 h. CONCLUSIONS: The (111)In-labeled IAC showed an improve tumor targeting kinetics with rapid accumulation and prolonged retention in the alpha(v)beta(3) receptor-positive tumor. This together with the rapid whole-body clearance pharmacokinetics warrants further studies on this IAC analog for molecular imaging of tumor-induced angiogenic vessels and various malignant human tumors expressing the receptor.


Subject(s)
Integrin alphaVbeta3/antagonists & inhibitors , Integrin alphaVbeta3/metabolism , Neoplasms/diagnostic imaging , Radiopharmaceuticals/pharmacokinetics , Sulfonamides/pharmacokinetics , beta-Alanine/analogs & derivatives , Animals , Humans , Indium Radioisotopes , Injections, Intraperitoneal , Isotope Labeling , Melanoma, Experimental/drug therapy , Mice , Mice, Nude , Molecular Mimicry , Neoplasm Transplantation , Neoplasms/metabolism , Tissue Distribution , Tomography, Emission-Computed, Single-Photon , beta-Alanine/pharmacokinetics
17.
Clin Cancer Res ; 13(9): 2722-7, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17473205

ABSTRACT

PURPOSE: To determine if pulsed-high intensity focused ultrasound (HIFU) could effectively serve as a source of hyperthermia with thermosensitive liposomes to enhance delivery and efficacy of doxorubicin in tumors. EXPERIMENTAL DESIGN: Comparisons in vitro and in vivo were carried out between non-thermosensitive liposomes (NTSL) and low temperature-sensitive liposomes (LTSL). Liposomes were incubated in vitro over a range of temperatures and durations, and the amount of doxorubicin released was measured. For in vivo experiments, liposomes and free doxorubicin were injected i.v. in mice followed by pulsed-HIFU exposures in s.c. murine adenocarcinoma tumors at 0 and 24 h after administration. Combinations of the exposures and drug formulations were evaluated for doxorubicin concentration and growth inhibition in the tumors. RESULTS: In vitro incubations simulating the pulsed-HIFU thermal dose (42 degrees C for 2 min) triggered release of 50% of doxorubicin from the LTSLs; however, no detectable release from the NTSLs was observed. Similarly, in vivo experiments showed that pulsed-HIFU exposures combined with the LTSLs resulted in more rapid delivery of doxorubicin as well as significantly higher i.t. concentration when compared with LTSLs alone or NTSLs, with or without exposures. Combining the exposures with the LTSLs also significantly reduced tumor growth compared with all other groups. CONCLUSIONS: Combining low-temperature heat-sensitive liposomes with noninvasive and nondestructive pulsed-HIFU exposures enhanced the delivery of doxorubicin and, consequently, its antitumor effects. This combination therapy could potentially produce viable clinical strategies for improved targeting and delivery of drugs for treatment of cancer and other diseases.


Subject(s)
Doxorubicin/administration & dosage , Drug Delivery Systems/methods , Hyperthermia, Induced/methods , Neoplasms/drug therapy , Ultrasonics , Animals , Cell Line, Tumor , Liposomes , Mice , Temperature
18.
Thromb Res ; 121(2): 193-202, 2007.
Article in English | MEDLINE | ID: mdl-17481699

ABSTRACT

INTRODUCTION: Thrombotic disease continues to account for significant morbidity and mortality. Ultrasound energy has been investigated as a potential primary and adjunctive treatment for thrombotic disease. We have previously shown that pulsed-high intensity focused ultrasound (HIFU) enhances thrombolysis induced by tissue plasminogen activator (tPA) in vitro, including describing the non-destructive mechanism by which tPA availability and consequent activity are increased. In this study we aimed to determine if the same effects could be achieved in vivo. MATERIALS AND METHODS: In this study, pulsed-HIFU exposures combined with tPA boluses were compared to treatment with tPA alone, HIFU alone and control in a novel in vivo clot model. Clots were formed in the rabbit marginal ear vein and verified using venography and infrared imaging. The efficacy of thrombolytic treatment was monitored via high resolution ultrasonography for 5 h post-treatment. The cross-sectional area of clots at 4 points along the vein was measured and normalized to the pre-treatment size. RESULTS: At 5 h the complete recanalization of clots treated with pulsed-HIFU and tPA was significantly different from the partial recanalization seen with tPA treatment alone. tPA treatment alone showed a significant decrease in clot versus control, where HIFU was not significantly different than control. Histological analysis of the vessel walls in the treated veins showed no apparent irreversible damage to endothelial cells or extravascular tissue. CONCLUSIONS: This study demonstrates that tPA mediated thrombolysis can be significantly enhanced when combined with non-invasive pulsed-HIFU exposures.


Subject(s)
Fibrinolytic Agents/therapeutic use , Thrombolytic Therapy/methods , Thrombosis/therapy , Tissue Plasminogen Activator/therapeutic use , Ultrasonic Therapy , Animals , Female , Models, Animal , Pilot Projects , Rabbits , Thrombosis/drug therapy , Treatment Outcome , Ultrasonics
19.
Bioconjug Chem ; 18(3): 821-8, 2007.
Article in English | MEDLINE | ID: mdl-17375899

ABSTRACT

Sulfhydryl selective reactions were explored to conjugate oligomers of a peptidomimetic integrin alphavbeta3 antagonist, 4-[2-(3,4,5,6-tetrahydropyrimidine-2-ylamino)ethyloxy]benzoyl-2-(S)-aminoethylsulfonylamino-beta-alanine (IA) to monoclonal antibody (MoAb) to increase integrin alphavbeta3 receptor-binding avidity. To generate sulfhydryl groups, N-succinimidyl-S-acetylthioacetate (SATA) was conjugated to both MoAb and IA. Sulfhydryl groups were then generated upon the deacetylation of the protecting acetyl group from the S-acetylthioacetato (ATA) moiety of MoAb-(ATA)n or IA-ATA with 0.02 M hydroxylamine in the presence of 1 mM EDTA at pH 7.2. The major focus was on optimizing the reaction concentrations, molar ratios, and reaction pH to conjugate high levels of IA-(A-SH) to MoAb-(A-SH)n without causing the inter- and intramolecular cross-linking of MoAb. Stepwise reactions of MoAb-(A-SH)n (15 microM MoAb) with a homobifunctional cross-linker, 1,8-bis(maleimido)diethylene glycol (BM[PEO]2) at a >50x molar excess to the -SH, followed by the reaction of the purified product MoAb-(A-S-succinimidomaleimido-[PEO]2)n with IA-(A-SH) at pH 7.2 afforded monomeric MoAb-(A-S-succinimido-[PEO]2-succinimido-S-A-IA)n with <10% high molecular weight oligomeric MoAb. Monomeric MoAb-(A-S-S-[PEO]2-S-S-A-IA)10 (MoAb-IA10) radiolabeled with 111In using 2-(p-isothiocyanatobenzyl)cyclohexyl-DTPA and with 125I using the Iodogen method showed >70% bindability to 0.4 microM alphavbeta3. When injected iv to nude mice with the receptor-positive M21 tumor, MoAb-IA10 radiolabeled with both 111In and 125I accumulated rapidly and was retained in the tumor for a 44 h period while the radioactivity cleared rapidly from the blood, thereby resulting in increasing tumor-to-blood ratios over time. The tumor uptake was similar between the 125I label and the 111In label for a 44 h period. In contrast, the blood radioactivity was lower, but liver and other organ uptakes were much higher for the 111In label than for the 125I. The 111In label produced higher tumor-to-blood ratios but much lower tumor-to-organ ratios than the 125I. The rapid blood clearance, a short peak tumor uptake time, and a low peak tumor uptake value with prolonged tumor retention of this macromolecule appear to support a hypothesis that MoAb-IA10 primarily binds to alphavbeta3 receptors on angiogenic vessels, but not on the tumor. This hypothesis was substantiated by the fluorescence microscopic analysis of FITC-MoAb-IA10, which showed that FITC-MoAb-IA10 outlined neovasculatures but not tumor cells at 4 and 21 h ex vivo. Additional proof was observed when blood vessels outlined with rhodamine-lectin, which specifically binds to blood vessels, were superimposable on neovasculatures outlined with FITC-MoAb-IA10.


Subject(s)
Angiogenesis Inhibitors/pharmacokinetics , Antibodies, Monoclonal/pharmacokinetics , Drug Carriers/pharmacokinetics , Integrin alphaVbeta3/antagonists & inhibitors , Neovascularization, Pathologic/metabolism , Sulfonamides/pharmacokinetics , beta-Alanine/analogs & derivatives , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/chemistry , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/chemistry , Biomimetic Materials/administration & dosage , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacokinetics , Cell Line, Tumor , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Fluorescein-5-isothiocyanate/chemistry , Humans , Indium Radioisotopes/chemistry , Iodine Radioisotopes/chemistry , Mice , Mice, Inbred Strains , Neoplasms/blood supply , Neoplasms/metabolism , Neovascularization, Pathologic/immunology , Succinimides/chemistry , Sulfides/chemistry , Sulfonamides/administration & dosage , Sulfonamides/chemistry , Tissue Distribution , Xenograft Model Antitumor Assays , beta-Alanine/administration & dosage , beta-Alanine/chemistry , beta-Alanine/pharmacokinetics
20.
Future Oncol ; 2(1): 111-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16556078

ABSTRACT

As the understanding of human cancer biology increases, new potential strategies for gene therapy are being proposed and evaluated. However, safe and efficient gene transfer continues to be the major hurdle for its implementation in the clinic. Preclinical studies have shown how pulsed-high intensity focused ultrasound (HIFU) exposures can be combined with different modes of administration (local, intravascular and systemic) to improve local delivery of genes and other therapeutic agents. Using image guidance, exposures are given, where short pulses of energy create predominantly mechanical/structural effects in the tissues as opposed to thermal ones. The result is an increase in both extravasation and interstitial diffusion of macromolecules, which occur non-destructively and reversibly. Ultrasound contrast agents can also be added, which enhance acoustic cavitation activity and consequently sonoporation. By being able to locally increase the uptake and expression of DNA, pulsed-HIFU holds much promise to further the use and applications of gene therapy for treating cancer and other pathological conditions.


Subject(s)
Genetic Therapy/methods , Ultrasonic Therapy/methods , Drug Delivery Systems , Hot Temperature/adverse effects , Humans , Neoplasms/therapy , Radiation
SELECTION OF CITATIONS
SEARCH DETAIL
...