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










Publication year range
1.
Article in English | MEDLINE | ID: mdl-34531932

ABSTRACT

BACKGROUND: In our earlier work, we identified microRNA-10b (miR10b) as a master regulator of the viability of metastatic tumor cells. This knowledge allowed us to design a miR10b-targeted therapeutic consisting of anti-miR10b and ultrasmall iron oxide magnetic nanoparticles (MN), termed MN-anti-miR10b. In mouse models of breast cancer, we demonstrated that MN-anti-miR10b caused durable regressions of established metastases with no evidence of systemic toxicity. As a first step towards translating MN-anti-miR10b for the treatment of metastatic breast cancer, we needed to determine if MN-anti-miR10b, which is so effective in mice, will also accumulate in human metastases. RESULTS: In this study, we devised a method to efficiently radiolabel MN-anti-miR10b with Cu-64 (64Cu) and evaluated the pharmacokinetics and biodistribution of the radiolabeled product at two different doses: a therapeutic dose, referred to as macrodose, corresponding to 64Cu-MN-anti-miR10b co-injected with non-labeled MN-anti-miR10b, and a tracer level dose of 64Cu-MN-anti-miR10b, referred to as microdose. In addition, we evaluated the uptake of 64Cu-MN-anti-miR10b by metastatic lesions using both in vivo and ex vivo positron emission tomography-magnetic resonance imaging (PET-MRI). A comparable distribution of the therapeutic was observed after administration of a microdose or macrodose. Uptake of the therapeutic by metastatic lymph nodes, lungs, and bone was also demonstrated by PET-MRI with a significantly higher PET signal than in the same organs devoid of metastatic lesions. CONCLUSION: Our results demonstrate that PET-MRI following a microdose injection of the agent will accurately reflect the innate biodistribution of the therapeutic. The tools developed in the present study lay the groundwork for the clinical testing of MN-anti-miR10b and other similar therapeutics in patients with cancer.

2.
Sci Rep ; 11(1): 2844, 2021 02 02.
Article in English | MEDLINE | ID: mdl-33531596

ABSTRACT

RNA interference represents one of the most appealing therapeutic modalities for cancer because of its potency, versatility, and modularity. Because the mechanism is catalytic and affects the expression of disease-causing antigens at the post-transcriptional level, only small amounts of therapeutic need to be delivered to the target in order to exert a robust therapeutic effect. RNA interference is also advantageous over other treatment modalities, such as monoclonal antibodies or small molecules, because it has a much broader array of druggable targets. Finally, the complementarity of the genetic code gives us the opportunity to design RNAi therapeutics using computational, rational approaches. Previously, we developed and tested an RNAi-targeted therapeutic, termed MN-anti-miR10b, which was designed to inhibit the critical driver of metastasis and metastatic colonization, miRNA-10b. We showed in animal models of metastatic breast cancer that MN-anti-miR10b accumulated into tumors and metastases in the lymph nodes, lungs, and bone, following simple intravenous injection. We also found that treatment incorporating MN-anti-miR10b was effective at inhibiting the emergence of metastases and could regress already established metastases in the lymph nodes, lungs, and bone. In the present study, we extend the application of MN-anti-miR10b to a model of breast cancer metastatic to the brain. We demonstrate delivery to the metastatic lesions and obtain evidence of a therapeutic effect manifested as inhibition of metastatic progression. This investigation represents an additional step towards translating similar RNAi-targeted therapeutics for the systemic treatment of metastatic disease.


Subject(s)
Brain Neoplasms/therapy , Breast Neoplasms/therapy , MicroRNAs/antagonists & inhibitors , RNA Interference , RNAi Therapeutics/methods , Animals , Brain Neoplasms/genetics , Brain Neoplasms/secondary , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Female , Humans , Mice , MicroRNAs/genetics , Xenograft Model Antitumor Assays
3.
Dalton Trans ; 49(40): 14088-14098, 2020 Oct 20.
Article in English | MEDLINE | ID: mdl-32970072

ABSTRACT

Synthesis and characterisation of a dithiadiaza chelator NSNS2A, as well as copper complexes thereof are reported in this paper. Solution structures of copper(i/ii) complexes were calculated using density functional theory (DFT) and validated by both NMR and EPR spectroscopy. DFT calculations revealed a switch in the orientation of tetragonal distortion upon protonation, which might be responsible for poor stability of the Cu(II)NSNS2A complex in aqueous media, whilst the same switch in tetragonal distortion was experimentally observed by changing the solvent. The chelator was radiolabeled with 64Cu and evaluated using PET/MRI in rats. Despite a favorable redox potential to stabilize the cuprous state in vivo, the 64Cu(II)NSNS2A complex showed suboptimal stability compared to its tetraazamacrocyclic analogue, 64Cu(TE2A), with a significant 64Cu uptake in the liver.


Subject(s)
Aza Compounds/chemistry , Chelating Agents/chemical synthesis , Coordination Complexes/chemical synthesis , Copper Radioisotopes/chemistry , Macrocyclic Compounds/chemical synthesis , Radiopharmaceuticals/chemical synthesis , Animals , Azurin/chemistry , Coordination Complexes/blood , Coordination Complexes/pharmacokinetics , Density Functional Theory , Electrochemical Techniques , Kidney , Liver , Magnetic Resonance Imaging/methods , Male , Molecular Conformation , Oxidation-Reduction , Positron-Emission Tomography/methods , Protein Binding , Radiopharmaceuticals/blood , Radiopharmaceuticals/pharmacology , Rats, Wistar , Solvents/chemistry , Staining and Labeling , Structure-Activity Relationship
4.
Angew Chem Int Ed Engl ; 59(4): 1474-1478, 2020 01 20.
Article in English | MEDLINE | ID: mdl-31750991

ABSTRACT

Gadolinium-based contrast agents (GBCAs) are used to provide diagnostic information in clinical magnetic resonance (MR) examinations. Gadolinium (Gd) has been detected in the brain, bone and skin of patients, months and years following GBCA administration, raising concerns about long term toxicity. Despite increased scrutiny, the concentration, chemical form and fate of the retained gadolinium species remain unknown. Importantly, the whole body biodistribution and organ clearance of GBCAs is poorly understood in humans. Gadolinium lacks suitable isotopes for nuclear imaging. We demonstrate that the yttrium-86 isotope can be used as a gadolinium surrogate. We show that Gd and their analogous Y complexes have similar properties both in solution and in vivo, and that yttrium-86 PET can be used to track the biodistribution of GBCAs over a two-day period.


Subject(s)
Gadolinium/chemistry , Magnetic Resonance Imaging/methods , Yttrium Radioisotopes/chemistry , Contrast Media , Humans
5.
Sci Rep ; 9(1): 4712, 2019 03 18.
Article in English | MEDLINE | ID: mdl-30886310

ABSTRACT

The recent past has seen impressive progress in the treatment of various malignancies using immunotherapy. One of the most promising approaches involves immune checkpoint inhibitors. However, the clinical results with these agents have demonstrated variability in the response. Pancreatic cancer, in particular, has proven resistant to initial immunotherapy approaches. Here, we describe an alternative strategy that relies on combining gemcitabine and a novel programmed death-ligand 1 (PD-L1) inhibitor, termed MN-siPDL1. MN-siPDL1 incorporates small interfering RNA against PD-L1 (siPDL1) conjugated to a magnetic nanocarrier (MN). We show that noninvasive magnetic resonance imaging (MRI) could be used to monitor therapeutic response. Combination therapy consisting of gemcitabine and MN-siPDL1 in a syngeneic murine pancreatic cancer model resulted in a significant reduction in tumor growth and an increase in survival. Following optimization, a 90% reduction in tumor volume was achieved 2 weeks after the beginning of treatment. Whereas 100% of the control animals had succumbed to their tumors by week 6 after the beginning of treatment, there was no mortality in the experimental group by week 5, and 67% of the experimental animals survived for 12 weeks. This method could provide therapeutic benefit against an intractable disease for which there are no effective treatments and which is characterized by a mere 1% 5-year survival.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Carcinoma, Pancreatic Ductal/drug therapy , Drug Carriers/chemistry , Immunotherapy/methods , Pancreatic Neoplasms/drug therapy , RNA, Small Interfering/administration & dosage , Animals , Antimetabolites, Antineoplastic/pharmacology , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Carcinoma, Pancreatic Ductal/diagnostic imaging , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor/transplantation , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Monitoring/methods , Female , Humans , Magnetic Resonance Imaging , Magnetite Nanoparticles/chemistry , Maximum Tolerated Dose , Mice , Pancreas/diagnostic imaging , Pancreas/drug effects , Pancreas/immunology , Pancreas/pathology , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , RNA Interference , RNA, Small Interfering/genetics , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Gemcitabine
6.
Quant Imaging Med Surg ; 8(2): 114-122, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29675353

ABSTRACT

BACKGROUND: Islet transplantation (Tx) represents the most promising therapy to restore normoglycemia in type 1 diabetes (T1D) patients to date. As significant islet loss has been observed after the procedure, there is an urgent need for developing strategies for monitoring transplanted islet grafts. In this report we describe for the first time the application of magnetic particle imaging (MPI) for monitoring transplanted islets in the liver and under the kidney capsule in experimental animals. METHODS: Pancreatic islets isolated from Papio hamadryas were labeled with superparamagnetic iron oxides (SPIOs) and used for either islet phantoms or Tx in the liver or under the kidney capsule of NOD scid mice. MPI was used to image and quantify islet phantoms and islet transplanted experimental animals post-mortem at 1 and 14 days after Tx. Magnetic resonance imaging (MRI) was used to confirm the presence of labeled islets in the liver and under the kidney capsule 1 day after Tx. RESULTS: MPI of labeled islet phantoms confirmed linear correlation between the number of islets and the MPI signal (R2=0.988). Post-mortem MPI performed on day 1 after Tx showed high signal contrast in the liver and under the kidney capsule. Quantitation of the signal supports islet loss over time, which is normally observed 2 weeks after Tx. No MPI signal was observed in control animals. In vivo MRI confirmed the presence of labeled islets/islet clusters in liver parenchyma and under the kidney capsule one day after Tx. CONCLUSIONS: Here we demonstrate that MPI can be used for quantitative detection of labeled pancreatic islets in the liver and under the kidney capsule of experimental animals. We believe that MPI, a modality with no depth attenuation and zero background tissue signal could be a suitable method for imaging transplanted islet grafts.

7.
J Biomed Opt ; 22(4): 40501, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28397959

ABSTRACT

Although the development of tumor-targeted fluorescent probes is a major area of investigation, it will be several years before these probes are realized for clinical use. Here, we report an approach that employs indocyanine-green (ICG), a clinically approved, nontargeted dye, in conjunction with fluorescence lifetime (FLT) detection to provide high accuracy for tumor-tissue identification in mouse models of subcutaneous human breast and brain tmors. The improved performance relies on the distinct FLTs of ICG within tumors versus tissue autofluorescence and is further aided by the well-known enhanced permeability and retention of ICG in tumors and the clearance of ICG from normal tissue several hours after intravenous injection. We demonstrate that FLT detection can provide more than 98% sensitivity and specificity, and a 10-fold reduction in error rates compared to intensity-based detection. Our studies suggest the significant potential of FLT-contrast for accurate tumor-tissue identification using ICG and other targeted probes under development, both for intraoperative imaging and for ex-vivo margin assessment of surgical specimens.


Subject(s)
Brain Neoplasms/diagnostic imaging , Breast Neoplasms/diagnostic imaging , Indocyanine Green/metabolism , Optical Imaging/methods , Animals , Fluorescence , Fluorescent Dyes/metabolism , Humans , Mice , Sensitivity and Specificity
8.
Sci Rep ; 7: 45060, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28322342

ABSTRACT

Treatment of stage IV metastatic breast cancer patients is limited to palliative options and represents an unmet clinical need. Here, we demonstrate that pharmacological inhibition of miRNA-10b - a master regulator of metastatic cell viability - leads to elimination of distant metastases in a mouse model of metastatic breast cancer. This was achieved using the miRNA-10b inhibitory nanodrug, MN-anti-miR10b, which consists of magnetic nanoparticles, conjugated to LNA-based miR-10b antagomirs. Intravenous injection of MN-anti-miR10b into mice bearing lung, bone, and brain metastases from breast cancer resulted in selective accumulation of the nanodrug in metastatic tumor cells. Weekly treatments of mice with MN-anti-miR-10b and low-dose doxorubicin resulted in complete regression of pre-existing distant metastases in 65% of the animals and a significant reduction in cancer mortality. These observations were supported by dramatic reduction in proliferation and increase in apoptosis in metastatic sites. On a molecular level, we observed a significant increase in the expression of HOXD10, which is a known target of miRNA-10b. These results represent first steps into the uncharted territory of therapy targeted to the metastatic niche.


Subject(s)
Breast Neoplasms/pathology , Breast Neoplasms/therapy , Models, Biological , Molecular Targeted Therapy , Animals , Apoptosis/genetics , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/etiology , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Doxorubicin/administration & dosage , Drug Delivery Systems , Female , Humans , Mice , MicroRNAs/administration & dosage , MicroRNAs/genetics , Nanomedicine , Nanoparticles/chemistry , Neoplasm Metastasis , Neoplasm Staging , Optical Imaging , Xenograft Model Antitumor Assays
9.
Diabetes Metab Res Rev ; 33(5)2017 07.
Article in English | MEDLINE | ID: mdl-28032446

ABSTRACT

BACKGROUND: The absence of reliable drug delivery systems to pancreatic islet cells hampers efficient treatment of type 1 diabetes. Nanoparticle delivery systems equipped with imaging capabilities could enable selective delivery to pancreatic islet cells. Biodistribution of nanoparticles is defined by several factors including the mode of administration, which determines accumulation in various organs. METHODS: In this study, we tested whether intrapancreatic ductal injection of magnetic nanoparticles would result in efficient cellular uptake by pancreatic islet cells. Dextran-coated iron oxide nanoparticles labeled with the near infrared fluorescent dye Cy5.5 were injected into the intrapancreatic ducts of streptozotocin-induced diabetic and healthy mice. To monitor the distribution of the nanoparticles, we performed in vivo magnetic resonance imaging followed by optical imaging and histology. RESULTS: Both imaging modalities demonstrated accumulation of the nanoparticles in the pancreas. However, histology revealed a high accumulation of nanoparticles in the insulin-producing cells in the pancreata of diabetic animals. By contrast, in nondiabetic controls, nanoparticles were mainly restricted to nonendocrine tissues. CONCLUSIONS: Our results demonstrate that pancreatic ductal injection accompanied by image guidance could serve as an alternative pathway for nanoparticle delivery. We expect to utilize this intraductal delivery method for theranostic applications in type 1 diabetes.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Drug Delivery Systems , Islets of Langerhans/metabolism , Magnetic Resonance Imaging/methods , Molecular Imaging/methods , Nanoparticles/administration & dosage , Pancreas/metabolism , Animals , Carbocyanines/chemistry , Diabetes Mellitus, Experimental/therapy , Female , Islets of Langerhans/pathology , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry , Pancreas/pathology , Tissue Distribution
10.
Int J Cancer ; 139(3): 712-8, 2016 08 01.
Article in English | MEDLINE | ID: mdl-26996122

ABSTRACT

The underglycosylated mucin 1 tumor antigen (uMUC1) is a biomarker that forecasts the progression of adenocarcinomas. In this study, we evaluated the utility of a dual-modality molecular imaging approach based on targeting uMUC1 for monitoring chemotherapeutic response in a transgenic murine model of pancreatic cancer (KCM triple transgenic mice). An uMUC1-specific contrast agent (MN-EPPT) was synthesized for use with magnetic resonance imaging (MRI) and fluorescence optical imaging. It consisted of dextran-coated iron oxide nanoparticles conjugated to the near infrared fluorescent dye Cy5.5 and to a uMUC1-specific peptide (EPPT). KCM triple transgenic mice were given gemcitabine as chemotherapy while control animals received saline injections following the same schedule. Changes in uMUC1 levels following chemotherapy were monitored using T2-weighted MRI and optical imaging before and 24 hr after injection of the MN-EPPT. uMUC1 expression in tumors from both groups was evaluated by histology and qRT-PCR. We observed that the average delta-T2 in the gemcitabine-treated group was significantly reduced compared to the control group indicating lower accumulation of MN-EPPT, and correspondingly, a lower level of uMUC1 expression. In vivo optical imaging confirmed the MRI findings. Fluorescence microscopy of pancreatic tumor sections showed a lower level of uMUC1 expression in the gemcitabine-treated group compared to the control, which was confirmed by qRT-PCR. Our data proved that changes in uMUC1 expression after gemcitabine chemotherapy could be evaluated using MN-EPPT-enhanced in vivo MR and optical imaging. These results suggest that the uMUC1-targeted imaging approach could provide a useful tool for the predictive assessment of therapeutic response.


Subject(s)
Antineoplastic Agents/pharmacology , Molecular Imaging , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/metabolism , Animals , Cell Line, Tumor , Contrast Media , Disease Models, Animal , Female , Humans , Magnetic Resonance Imaging , Male , Mice , Mice, Transgenic , Molecular Imaging/methods , Mucin-1/metabolism , Optical Imaging/methods , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Reproducibility of Results , Treatment Outcome
11.
Mol Imaging Biol ; 18(1): 70-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25987466

ABSTRACT

PURPOSE: The development of tools for the analysis of microRNA (miRNA) function in tumors can advance our diagnostic and prognostic capabilities. Here, we describe the development of technology for the profiling of miRNA expression in the tumors of live animals. PROCEDURES: The approach is based on miRNA nanosensors consisting of sensor oligonucleotides conjugated to magnetic nanoparticles for systemic delivery. Feasibility was demonstrated for the detection of miR-10b, implicated in epithelial to mesenchymal transition and the development of metastasis. The miR-10b nanosensor was tested in vivo in two mouse models of cancer. In the first model, mice were implanted subcutaneously with MDA-MB-231-luc-D3H2LN tumors, in which miR-10b was inhibited. In the second model, mice were implanted bilaterally with metastatic MDA-MB-231 and nonmetastatic MCF-7 cells. The nanosensors were injected intravenously, and fluorescence intensity in the tumors was monitored over time. RESULTS: We showed that the described nanosensors are capable of discriminating between tumors based on their expression of miR-10b. Radiant efficiency was higher in the miR-10b-active tumors than in the miR-10b-inhibited tumors and in the MDA-MB-231 tumors relative to the MCF-7 tumors. CONCLUSIONS: The described technology provides an important tool that could be used to answer questions about microRNA function in cancer.


Subject(s)
Biosensing Techniques/methods , MicroRNAs/metabolism , Nanoparticles/chemistry , Neoplasms/genetics , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Fluorescence , Gene Expression Regulation, Neoplastic , Humans , Mice, Nude , MicroRNAs/genetics , Oligonucleotides/metabolism , Xenograft Model Antitumor Assays
12.
Cancer Res ; 75(20): 4407-15, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26359455

ABSTRACT

The therapeutic promise of microRNA (miRNA) in cancer has yet to be realized. In this study, we identified and therapeutically exploited a new role for miR-10b at the metastatic site, which links its overexpression to tumor cell viability and proliferation. In the protocol developed, we combined a miR-10b-inhibitory nanodrug with low-dose anthracycline to achieve complete durable regressions of metastatic disease in a murine model of metastatic breast cancer. Mechanistic investigations suggested a potent antiproliferative, proapoptotic effect of the nanodrug in the metastatic cells, potentiated by a cell-cycle arrest produced by administration of the low-dose anthracycline. miR-10b was overexpressed specifically in cells with high metastatic potential, suggesting a role for this miRNA as a metastasis-specific therapeutic target. Taken together, our results implied the existence of pathways that regulate the viability and proliferation of tumor cells only after they have acquired the ability to grow at distant metastatic sites. As illustrated by miR-10b targeting, such metastasis-dependent apoptotic pathways would offer attractive targets for further therapeutic exploration.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Breast Neoplasms/genetics , Doxorubicin/administration & dosage , MicroRNAs/genetics , Nanoparticles , Animals , Apoptosis/genetics , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Cell Cycle/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Disease Models, Animal , Female , Gene Knockout Techniques , Humans , Mice , Neoplasm Metastasis , Phenotype , Tumor Burden/drug effects , Tumor Burden/genetics , Xenograft Model Antitumor Assays
13.
Proc Natl Acad Sci U S A ; 112(31): 9734-9, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26199414

ABSTRACT

Near-infrared fluorescence (NIRF) molecular imaging has been widely applied to monitoring therapy of cancer and other diseases in preclinical studies; however, this technology has not been applied successfully to monitoring therapy for Alzheimer's disease (AD). Although several NIRF probes for detecting amyloid beta (Aß) species of AD have been reported, none of these probes has been used to monitor changes of Aßs during therapy. In this article, we demonstrated that CRANAD-3, a curcumin analog, is capable of detecting both soluble and insoluble Aß species. In vivo imaging showed that the NIRF signal of CRANAD-3 from 4-mo-old transgenic AD (APP/PS1) mice was 2.29-fold higher than that from age-matched wild-type mice, indicating that CRANAD-3 is capable of detecting early molecular pathology. To verify the feasibility of CRANAD-3 for monitoring therapy, we first used the fast Aß-lowering drug LY2811376, a well-characterized beta-amyloid cleaving enzyme-1 inhibitor, to treat APP/PS1 mice. Imaging data suggested that CRANAD-3 could monitor the decrease in Aßs after drug treatment. To validate the imaging capacity of CRANAD-3 further, we used it to monitor the therapeutic effect of CRANAD-17, a curcumin analog for inhibition of Aß cross-linking. The imaging data indicated that the fluorescence signal in the CRANAD-17-treated group was significantly lower than that in the control group, and the result correlated with ELISA analysis of brain extraction and Aß plaque counting. It was the first time, to our knowledge, that NIRF was used to monitor AD therapy, and we believe that our imaging technology has the potential to have a high impact on AD drug development.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Drug Monitoring , Molecular Imaging/methods , Spectroscopy, Near-Infrared , Animals , Benzothiazoles , Carbon-13 Magnetic Resonance Spectroscopy , Disease Models, Animal , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , Humans , Mice, Transgenic , Photons , Presenilin-1/metabolism , Proton Magnetic Resonance Spectroscopy , Spectrometry, Fluorescence , Thiazoles/metabolism , Tissue Extracts , Titrimetry
14.
Diabetes ; 63(5): 1465-74, 2014 May.
Article in English | MEDLINE | ID: mdl-24458362

ABSTRACT

Noninvasive assessment of pancreatic ß-cell mass would tremendously aid in managing type 1 diabetes (T1D). Toward this goal, we synthesized an exendin-4 conjugated magnetic iron oxide-based nanoparticle probe targeting glucagon-like peptide 1 receptor (GLP-1R), which is highly expressed on the surface of pancreatic ß-cells. In vitro studies in ßTC-6, the ß-cell line, showed specific accumulation of the targeted probe (termed MN-Ex10-Cy5.5) compared with nontargeted (termed MN-Cy5.5). In vivo magnetic resonance imaging showed a significant transverse relaxation time (T2) shortening in the pancreata of mice injected with the MN-Ex10-Cy5.5 probe compared with control animals injected with the nontargeted probe at 7.5 and 24 h after injection. Furthermore, ΔT2 of the pancreata of prediabetic NOD mice was significantly higher than that of diabetic NOD mice after the injection of MN-Ex10-Cy5.5, indicating the decrease of probe accumulation in these animals due to ß-cell loss. Of note, ΔT2 of prediabetic and diabetic NOD mice injected with MN-Cy5.5 was not significantly changed, reflecting the nonspecific mode of accumulation of nontargeted probe. We believe our results point to the potential for using this agent for monitoring the disease development and response of T1D to therapy.


Subject(s)
Diabetes Mellitus, Type 1/pathology , Islets of Langerhans/pathology , Magnetite Nanoparticles , Pancreas/pathology , Receptors, Glucagon/metabolism , Animals , Cell Line, Tumor , Cell Survival , Diabetes Mellitus, Type 1/metabolism , Female , Glucagon-Like Peptide-1 Receptor , Insulinoma/metabolism , Insulinoma/pathology , Islets of Langerhans/metabolism , Magnetic Resonance Imaging , Mice , Mice, Inbred NOD , Pancreas/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology
15.
Int J Cancer ; 134(7): 1758-66, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24114765

ABSTRACT

The clinical management of cancer reflects a balance between treatment efficacy and toxicity. While typically, combination therapy improves response rate and time to progression compared with sequential monotherapy, it causes increased toxicity. Consequently, in cases of advanced cancer, emerging guidelines recommend sequential monotherapy, as a means to enhance quality of life. An alternative approach that could overcome nonspecific toxicity while retaining therapeutic efficacy, involves the combination of chemotherapy with targeted therapy. In the current study, we tested the hypothesis that combination therapy targeting survivin (BIRC5) and low-dose doxorubicin (Dox) will show enhanced therapeutic potential in the treatment of cancer, as compared to monotherapy with Dox. We demonstrate in both in vitro and in vivo models of breast cancer that combination therapy with a low dose of Dox and an anti-survivin siRNA nanodrug (MN-siBIRC5) is superior to mono-therapy with either low- or high-dose Dox alone. Importantly, therapeutic efficacy showed prominent sequence dependence. Induction of apoptosis was observed only when the cells were treated with Dox followed by MN-siBIRC5, whereas the reverse sequence abrogated the benefit of the drug combination. In vivo, confirmation of successful sequence dependent combination therapy was demonstrated in a murine xenograft model of breast cancer. Finally, to determine if the observed effect is not limited to breast cancer, we extended our studies to a murine xenograft model of pancreatic adenocarcinoma and found similar outcomes as shown for breast cancer.


Subject(s)
Adenocarcinoma/drug therapy , Doxorubicin/pharmacology , Inhibitor of Apoptosis Proteins/genetics , Nanoparticles/administration & dosage , RNA, Small Interfering/genetics , Adenocarcinoma/genetics , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Combined Modality Therapy/methods , Female , Humans , Mice , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Survivin
16.
Radiology ; 266(3): 822-30, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23264346

ABSTRACT

PURPOSE: To detect adoptively transferred immune attack in a mouse model of islet cell transplantation by using a long-circulating paramagnetic T1 contrast agent, a protected graft copolymer (PGC) that is covalently linked to gadolinium-diethylenetriaminepentaacetic acid with fluorescein isothiocyanate (Gd-DTPA-F), which accumulates in the sites of inflammation that are characterized by vascular disruption. MATERIALS AND METHODS: All animal experiments were performed in compliance with institutional guidelines and approved by the subcommittee on research animal care. Six nonobese diabetic severe combined immunodeficiency mice received transplanted human islet cells under the kidney capsule and adoptively transferred 5 × 10(6) splenocytes from 6-week-old nonobese diabetic mice. These mice also served as control subjects for comparison of pre- and postadoptive transfer MR imaging results. Mice that received phosphate-buffered saline solution only were included as nonadoptive-transfer control subjects (n = 2). In vivo magnetic resonance (MR) imaging was performed before and 17 hours after intravenous injections of PGC-Gd-DTPA-F, followed by histologic examination. Statistical differences were analyzed by means of a paired Student t test and repeated two-way analysis of variance. RESULTS: MR imaging results showed significantly greater accumulation of PGC-Gd-DTPA-F in the graft area after immune attack initiated by adoptive transfer of splenocytes compared with that of the same area before the transfer (T1, 137.2 msec ± 39.3 and 239.5 msec ± 17.6, respectively; P < .001). These results were confirmed at histologic examination, which showed considerable leakage of the contrast agent into the islet cell interstitium. CONCLUSION: PGC-Gd-DTPA-F-enhanced MR imaging allows for the in vivo assessment of vascular damage of the graft T cell challenge.


Subject(s)
Dextrans , Fluorescein-5-isothiocyanate/analogs & derivatives , Gadolinium DTPA , Graft Rejection/etiology , Graft Rejection/pathology , Islets of Langerhans Transplantation/adverse effects , Islets of Langerhans Transplantation/pathology , Magnetic Resonance Imaging/methods , Animals , Cells, Cultured , Contrast Media/administration & dosage , Dextrans/administration & dosage , Diabetes Mellitus/pathology , Diabetes Mellitus/surgery , Fluorescein-5-isothiocyanate/administration & dosage , Gadolinium DTPA/administration & dosage , Humans , Mice , Reproducibility of Results , Sensitivity and Specificity
17.
Int J Cancer ; 132(8): 1860-7, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23015160

ABSTRACT

The ability to monitor breast cancer initiation and progression on the molecular level would provide an effective tool for early diagnosis and therapy. In the present study, we focused on the underglycosylated MUC-1 tumor antigen (uMUC-1), which is directly linked to tumor progression from pre-malignancy to advanced malignancy in breast cancer and has been identified as the independent predictor of local recurrence and tumor response to chemotherapy. We investigated whether changes in uMUC-1 expression during tumor development and therapeutic intervention could be monitored non-invasively using molecular imaging approach with the uMUC-1-specific contrast agent (MN-EPPT) detectable by magnetic resonance and fluorescence optical imaging. This was done in mice that express human uMUC-1 tumor antigen (MMT mice) and develop spontaneous mammary carcinoma in a stage-wise fashion. After the injection of MN-EPPT there was a significant reduction in average T2 relaxation times of the mammary fat pad between pre-malignancy and cancer. In addition, T2 relaxation times were already altered at pre-malignant state in these mice compared to non-tumor bearing mice. This indicated that targeting uMUC-1 could be useful for detecting pre-malignant transformation in the mammary fat pad. We also probed changes in uMUC-1 expression with MN-EPPT during therapy with doxorubicin (Dox). We observed that tumor delta-T2s were significantly reduced by treatment with Dox indicating lower accumulation of MN-EPPT. This correlated with a lower level of MUC-1 expression in the Dox-treated tumors, as confirmed by immunoblotting. Our study could provide a very sensitive molecular imaging approach for monitoring tumor progression and therapeutic response.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/pathology , Doxorubicin/therapeutic use , Molecular Imaging , Mucin-1/metabolism , Animals , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Disease Models, Animal , Disease Progression , Down-Regulation , Female , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic
18.
Diabetes ; 61(12): 3247-54, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22923469

ABSTRACT

Islet transplantation has recently emerged as an acceptable clinical modality for restoring normoglycemia in patients with type 1 diabetes mellitus (T1DM). The long-term survival and function of islet grafts is compromised by immune rejection-related factors. Downregulation of factors that mediate immune rejection using RNA interference holds promise for improving islet graft resistance to damaging factors after transplantation. Here, we used a dual-purpose therapy/imaging small interfering (si)RNA magnetic nanoparticle (MN) probe that targets ß(2) microglobulin (B2M), a key component of the major histocompatibility class I complex (MHC I). In addition to serving as a siRNA carrier, this MN-siB2M probe enables monitoring of graft persistence noninvasively using magnetic resonance imaging (MRI). Human islets labeled with these MNs before transplantation into B2M (null) NOD/scid mice showed significantly improved preservation of graft volume starting at 2 weeks, as determined by longitudinal MRI in an adoptive transfer model (P < 0.05). Furthermore, animals transplanted with MN-siB2M-labeled islets demonstrated a significant delay of up to 23.8 ± 4.8 days in diabetes onset after the adoptive transfer of T cells relative to 6.5 ± 4.5 days in controls. This study demonstrated that our approach could protect pancreatic islet grafts from immune rejection and could potentially be applied to allotransplantation and prevention of the autoimmune recurrence of T1DM in islet transplantation or endogenous islets.


Subject(s)
Graft Rejection/prevention & control , Islets of Langerhans Transplantation/immunology , Islets of Langerhans Transplantation/methods , RNA, Small Interfering/physiology , Animals , Humans , Major Histocompatibility Complex/genetics , Mice , Mice, Inbred NOD , Mice, SCID , RNA, Small Interfering/genetics
19.
Gen Dent ; 56(7): 629-34, 2008.
Article in English | MEDLINE | ID: mdl-19014021

ABSTRACT

Low level laser therapy (LLLT) uses light energy, in the form of adenosine triphosphate (ATP), to elicit biological responses in the body. The increased cellular energy and changes in the cell membrane permeability result in pain relief, wound healing, muscle relaxation, immune system modulation, and nerve regeneration. This article investigates the clinical effects of LLLT and explains how it can be applied in the dental field.


Subject(s)
Dental Care , Low-Level Light Therapy , Adenosine Triphosphate/radiation effects , Arthritis/radiotherapy , Cell Membrane Permeability/radiation effects , Dental Implants , Dental Restoration, Permanent , Dentin Sensitivity/radiotherapy , Facial Pain/radiotherapy , Humans , Immunity/radiation effects , Low-Level Light Therapy/methods , Muscle Relaxation/radiation effects , Muscle, Skeletal/radiation effects , Nerve Regeneration/radiation effects , Orthodontics, Corrective , Osteoarthritis/radiotherapy , Pain/radiotherapy , Pain, Postoperative/prevention & control , Periodontal Diseases/radiotherapy , Safety , Temporomandibular Joint Disorders/radiotherapy , Tooth Diseases/radiotherapy , Tooth Extraction , Wound Healing/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...