Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Pharmaceuticals (Basel) ; 17(1)2023 Dec 21.
Article in English | MEDLINE | ID: mdl-38275997

ABSTRACT

[64Cu]Cu-diacetyl-bis(N4-methylthiosemicarbazone) ([64Cu]Cu-ATSM) is a radioactive hypoxia-targeting therapeutic agent being investigated in clinical trials for malignant brain tumors. For the quality management of [64Cu]Cu-ATSM, understanding trace metal impurities' effects on the chelate formation of 64Cu and ATSM is important. In this study, we conducted coordination chemistry studies on metal-ATSM complexes. First, the effects of nonradioactive metal ions (Cu2+, Ni2+, Zn2+, and Fe2+) on the formation of [64Cu]Cu-ATSM were evaluated. When the amount of Cu2+ or Ni2+ added was 1.2 mol or 288 mol, equivalent to ATSM, the labeling yield of [64Cu]Cu-ATSM fell below 90%. Little effect was observed even when excess amounts of Zn2+ or Fe2+ were added to the ATSM. Second, these metals were reacted with ATSM, and chelate formation was measured using ultraviolet-visible (UV-Vis) absorption spectra. UV-Vis spectra showed a rapid formation of Cu2+ and the ATSM complex upon mixing. The rate of chelate formation by Ni2+ and ATSM was lower than that by Cu-ATSM. Zn2+ and Fe2+ showed much slower reactions with the ATSM than Ni2+. Trace amounts of Ni2+, Zn2+, and Fe2+ showed little effect on [64Cu]Cu-ATSM' quality, while the concentration of impurity Cu2+ must be controlled. These results can provide process management tools for radiopharmaceuticals.

2.
Biomaterials ; 51: 278-289, 2015 May.
Article in English | MEDLINE | ID: mdl-25771018

ABSTRACT

Anti-cancer drug development typically utilizes high-throughput screening with two-dimensional (2D) cell culture. However, 2D culture induces cellular characteristics different from tumors in vivo, resulting in inefficient drug development. Here, we report an innovative high-throughput screening system using nanoimprinting 3D culture to simulate in vivo conditions, thereby facilitating efficient drug development. We demonstrated that cell line-based nanoimprinting 3D screening can more efficiently select drugs that effectively inhibit cancer growth in vivo as compared to 2D culture. Metabolic responses after treatment were assessed using positron emission tomography (PET) probes, and revealed similar characteristics between the 3D spheroids and in vivo tumors. Further, we developed an advanced method to adopt cancer cells from patient tumor tissues for high-throughput drug screening with nanoimprinting 3D culture, which we termed Cancer tissue-Originated Uniformed Spheroid Assay (COUSA). This system identified drugs that were effective in xenografts of the original patient tumors. Nanoimprinting 3D spheroids showed low permeability and formation of hypoxic regions inside, similar to in vivo tumors. Collectively, the nanoimprinting 3D culture provides easy-handling high-throughput drug screening system, which allows for efficient drug development by mimicking the tumor environment. The COUSA system could be a useful platform for drug development with patient cancer cells.


Subject(s)
Cell Culture Techniques/methods , Drug Design , Drug Screening Assays, Antitumor/methods , Nanotechnology/methods , Tumor Microenvironment , Animals , Antineoplastic Agents/pharmacology , Biological Assay , Cell Hypoxia/drug effects , Cell Line, Tumor , Humans , Leupeptins/pharmacology , Male , Mice, Inbred BALB C , Mice, Nude , Neoplasms/pathology , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Tumor Microenvironment/drug effects
3.
Basic Res Cardiol ; 110(1): 455, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25449896

ABSTRACT

Brain natriuretic peptide (BNP) contributes to heart formation during embryogenesis. After birth, despite a high number of studies aimed at understanding by which mechanism(s) BNP reduces myocardial ischemic injury in animal models, the actual role of this peptide in the heart remains elusive. In this study, we asked whether BNP treatment could modulate the proliferation of endogenous cardiac progenitor cells (CPCs) and/or their differentiation into cardiomyocytes. CPCs expressed the NPR-A and NPR-B receptors in neonatal and adult hearts, suggesting their ability to respond to BNP stimulation. BNP injection into neonatal and adult unmanipulated mice increased the number of newly formed cardiomyocytes (neonatal: +23 %, p = 0.009 and adult: +68 %, p = 0.0005) and the number of proliferating CPCs (neonatal: +142 %, p = 0.002 and adult: +134 %, p = 0.04). In vitro, BNP stimulated CPC proliferation via NPR-A and CPC differentiation into cardiomyocytes via NPR-B. Finally, as BNP might be used as a therapeutic agent, we injected BNP into mice undergoing myocardial infarction. In pathological conditions, BNP treatment was cardioprotective by increasing heart contractility and reducing cardiac remodelling. At the cellular level, BNP stimulates CPC proliferation in the non-infarcted area of the infarcted hearts. In the infarcted area, BNP modulates the fate of the endogenous CPCs but also of the infiltrating CD45(+) cells. These results support for the first time a key role for BNP in controlling the progenitor cell proliferation and differentiation after birth. The administration of BNP might, therefore, be a useful component of therapeutic approaches aimed at inducing heart regeneration.


Subject(s)
Heart/drug effects , Myocardium/cytology , Natriuretic Peptide, Brain/pharmacology , Stem Cells/drug effects , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/analysis , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/drug therapy , Myocytes, Cardiac/physiology , Receptors, Atrial Natriuretic Factor/physiology , Stem Cells/cytology , Stem Cells/physiology , Transcription Factors/analysis
4.
Reprod Biol Endocrinol ; 12: 64, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-25012822

ABSTRACT

BACKGROUND: C-type natriuretic peptide (CNP) signaling through its receptor natriuretic peptide receptor B (NPR-B) is a key molecule for mammalian reproduction, and known to play important roles in female fertility. However, the function of these peptides in mouse male reproduction remains largely unknown. To determine the role of CNP/NPR-B signaling in male reproduction we investigated phenotype of Npr2-deficient short-limbed-dwarfism (Npr2(slw/slw)) mice, which have been shown to have gastrointestinal (GI) abnormalities. FINDINGS: In homozygous Npr2(slw/slw) mice, spermatogenesis is developmentally delayed at both 2 and 4 weeks of age, with vacuolation and degenerating apoptotic germ cells being observed at 3 weeks age. However, the adult Npr2(slw/slw) mice exhibited apparently normal spermatogenesis, albeit with some aberrant spermatids, suggesting that developmental delay was overcome. In addition, the adult Npr2(slw/slw) mice showed abnormal penile morphology (paraphimosis). CONCLUSIONS: The potential role of CNP signaling via the NPR-B receptor in male fertility appears to be mediated not through germ-cell development, but may be through maintenance of normal penile function.


Subject(s)
Erectile Dysfunction/etiology , Infertility, Male/metabolism , Paraphimosis/etiology , Penile Erection , Penis/metabolism , Receptors, Atrial Natriuretic Factor/metabolism , Animals , Apoptosis , Crosses, Genetic , Homozygote , Infertility, Male/genetics , Infertility, Male/pathology , Infertility, Male/physiopathology , Male , Mice, Inbred Strains , Mice, Mutant Strains , Mutation , Penis/physiopathology , Receptors, Atrial Natriuretic Factor/genetics , Spermatids/metabolism , Spermatids/pathology , Spermatogenesis , Spermatozoa/metabolism , Spermatozoa/pathology , Testicular Diseases/etiology , Vacuoles
5.
PLoS One ; 9(1): e86996, 2014.
Article in English | MEDLINE | ID: mdl-24466309

ABSTRACT

PURPOSE: (64)Cu-diacetyl-bis (N (4)-methylthiosemicarbazone) ((64)Cu-ATSM) is a promising theranostic agent that targets hypoxic regions in tumors related to malignant characteristics. Its diagnostic usefulness has been recognized in clinical studies. Internal radiotherapy (IRT) with (64)Cu-ATSM is reportedly effective in preclinical studies; however, for clinical applications, improvements to reduce radiation exposure in non-target organs, particularly the liver, are required. We developed a strategy to reduce radiation doses to critical organs while preserving tumor radiation doses by controlled administration of copper chelator penicillamine during (64)Cu-ATSM IRT. METHODS: Biodistribution was evaluated in HT-29 tumor-bearing mice injected with (64)Cu-ATSM (185 kBq) with or without oral penicillamine administration. The appropriate injection interval between (64)Cu-ATSM and penicillamine was determined. Then, the optimal penicillamine administration schedule was selected from single (100, 300, and 500 mg/kg) and fractionated doses (100 mg/kg×3 at 1- or 2-h intervals from 1 h after (64)Cu-ATSM injection). PET imaging was performed to confirm the effect of penicillamine with a therapeutic (64)Cu-ATSM dose (37 MBq). Dosimetry analysis was performed to estimate human absorbed doses. RESULTS: Penicillamine reduced (64)Cu accumulation in the liver and small intestine. Tumor uptake was not affected by penicillamine administration at 1 h after (64)Cu-ATSM injection, when radioactivity was almost cleared from the blood and tumor uptake had plateaued. Of the single doses, 300 mg/kg was most effective. Fractionated administration at 2-h intervals further decreased liver accumulation at later time points. PET indicated that penicillamine acts similarly with the therapeutic (64)Cu-ATSM dose. Dosimetry demonstrated that appropriately scheduled penicillamine administration reduced radiation doses to critical organs (liver, ovaries, and red marrow) below tolerance levels. Laxatives reduced radiation doses to the large intestine. CONCLUSIONS: We developed a novel strategy to reduce radiation exposure in critical organs during (64)Cu-ATSM IRT, thus promoting its clinical applications. This method could be beneficial for other (64)Cu-labeled compounds.


Subject(s)
Colonic Neoplasms/radiotherapy , Liver/drug effects , Organometallic Compounds/adverse effects , Penicillamine/pharmacology , Radiation Injuries/prevention & control , Radiation-Protective Agents/pharmacology , Thiosemicarbazones/adverse effects , Animals , Coordination Complexes , Delayed-Action Preparations , Dose-Response Relationship, Drug , Mice , Organometallic Compounds/therapeutic use , Penicillamine/administration & dosage , Positron-Emission Tomography , Radiation-Protective Agents/administration & dosage , Thiosemicarbazones/therapeutic use
6.
Eur J Pharm Biopharm ; 86(3): 478-86, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24316338

ABSTRACT

(64)Cu-cyclam-RAFT-c(-RGDfK-)4, an αVß3 integrin-targeting tetrameric cyclic RGD peptide probe, is a potential theranostic compound for positron emission tomography (PET) of tumor angiogenesis and for internal radiotherapy owing to the multiple decay modes of (64)Cu. Since kidneys are dose-limiting organs in internal radiotherapy, we aimed to reduce the renal accumulation of (64)Cu-cyclam-RAFT-c(-RGDfK-)4 by co-injection with Gelofusine (GF), a succinylated gelatin solution, and/or L-lysine (Lys), and to explore, for the first time, the related mechanisms using the noninvasive and quantitative PET imaging technology. Biodistribution assays, dynamic and static PET scans, and metabolism studies with radio-thin-layer chromatography (radio-TLC) were performed in healthy or αVß3-positive tumor-bearing mice. In the results, co-injection with GF markedly reduced the renal uptake and slightly increased the tumor uptake of (64)Cu-cyclam-RAFT-c(-RGDfK-)4. L-Lysine alone had no effect on the probe biodistribution, but the combined use of Lys and GF tended to enhance the effect of GF. Dynamic PET and metabolite analysis by radio-TLC highly revealed that GF blocks the renal reabsorption of (64)Cu-cyclam-RAFT-c(-RGDfK-)4, but does not interfere with its metabolism and excretion. In conclusion, administration of GF and Lys is a useful strategy for kidney protection in (64)Cu-cyclam-RAFT-c(-RGDfK-)4-based internal radiotherapy.


Subject(s)
Coordination Complexes/metabolism , Gelatin/metabolism , Kidney/diagnostic imaging , Kidney/metabolism , Lysine/metabolism , Peptides, Cyclic/metabolism , Positron-Emission Tomography/methods , Succinates/metabolism , Animals , Cell Line, Tumor , Drug Combinations , Drug Interactions , Female , Gelatin/administration & dosage , Humans , Kidney/drug effects , Lysine/administration & dosage , Mice , Mice, Inbred BALB C , Mice, Nude , Succinates/administration & dosage , Tissue Distribution/drug effects , Tissue Distribution/physiology , Xenograft Model Antitumor Assays/methods
7.
Mol Imaging ; 12(6): 376-87, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23981783

ABSTRACT

The purpose of this study was to develop a clinically relevant orthotopic xenotransplantation model of pancreatic cancer and to perform a preclinical evaluation of a new positron emission tomography (PET) imaging probe, 64Cu-labeled cyclam-RAFT-c(-RGDfK-)4 peptide (64Cu-RAFT-RGD), using this model. Varying degrees of αvß3 integrin expression in several human pancreatic cancer cell lines were examined by flow cytometry and Western blotting. The cell line BxPC-3, which is stably transfected with a red fluorescence protein (RFP), was used for surgical orthotopic implantation. Orthotopic xenograft was established in the pancreas of recipient nude mice. An in vivo probe biodistribution and receptor blocking study, preclinical PET imaging coregistered with contrast-enhanced computed tomography (CECT) comparing 64Cu-RAFT-RGD and ¹8F-fluoro-2-deoxy-d-glucose (¹8F-FDG) accumulation in tumor, postimaging autoradiography, and histologic and immunohistochemical examinations were done. Biodistribution evaluation with a blocking study confirmed that efficient binding of probe to tumor is highly αvß3 integrin specific. 64Cu-RAFT-RGD PET combined with CECT provided for precise and easy detection of cancer lesions. Autoradiography, histologic, and immunohistochemical examinations confirmed the accumulation of 64Cu-RAFT-RGD in tumor versus nontumor tissues. In comparative PET studies, 64Cu-RAFT-RGD accumulation provided better tumor contrast to background than ¹8F-FDG. Our results suggest that 64Cu-RAFT-RGD PET imaging is potentially applicable for the diagnosis of αvß3 integrin-expressing pancreatic tumors.


Subject(s)
Coordination Complexes , Integrin alphaVbeta3/analysis , Pancreatic Neoplasms/diagnostic imaging , Peptides, Cyclic , Positron-Emission Tomography/methods , Radiopharmaceuticals , X-Ray Microtomography/methods , Animals , Cell Line, Tumor , Coordination Complexes/pharmacokinetics , Copper Radioisotopes , Female , Heterografts , Histocytochemistry , Humans , Integrin alphaVbeta3/biosynthesis , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Peptides, Cyclic/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Reproducibility of Results , Tissue Distribution
8.
PLoS One ; 8(5): e64570, 2013.
Article in English | MEDLINE | ID: mdl-23741342

ABSTRACT

Fatty acid synthase (FASN) expression is elevated in several cancers, and this over-expression is associated with poor prognosis. Inhibitors of FASN, such as orlistat, reportedly show antitumor effects against cancers that over-express FASN, making FASN a promising therapeutic target. However, large variations in FASN expression levels in individual tumors have been observed, and methods to predict FASN-targeted therapy outcome before treatment are required to avoid unnecessary treatment. In addition, how FASN inhibition affects tumor progression remains unclear. Here, we showed the method to predict FASN-targeted therapy outcome using radiolabeled acetate uptake and presented mechanisms of FASN inhibition with human prostate cancer cell lines, to provide the treatment strategy of FASN-targeted therapy. We revealed that tumor uptake of radiolabeled acetate reflected the FASN expression levels and sensitivity to FASN-targeted therapy with orlistat in vitro and in vivo. FASN-targeted therapy was noticeably effective against tumors with high FASN expression, which was indicated by high acetate uptake. To examine mechanisms, we established FASN knockdown prostate cancer cells by transduction of short-hairpin RNA against FASN and investigated the characteristics by analyses on morphology and cell behavior and microarray-based gene expression profiling. FASN inhibition not only suppressed cell proliferation but prevented pseudopodia formation and suppressed cell adhesion, migration, and invasion. FASN inhibition also suppressed genes involved in production of intracellular second messenger arachidonic acid and androgen hormones, both of which promote tumor progression. Collectively, our data demonstrated that uptake of radiolabeled acetate is a useful predictor of FASN-targeted therapy outcome. This suggests that [1-(11)C]acetate positron emission tomography (PET) could be a powerful tool to accomplish personalized FASN-targeted therapy by non-invasive visualization of tumor acetate uptake and selection of responsive tumors. FASN-targeted therapy could be an effective treatment to suppress multiple steps related to tumor progression in prostate cancers selected by [1-(11)C]acetate PET.


Subject(s)
Acetic Acid , Adenocarcinoma/drug therapy , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Fatty Acid Synthase, Type I/metabolism , Lactones/pharmacology , Prostatic Neoplasms/drug therapy , Adenocarcinoma/diagnosis , Adenocarcinoma/enzymology , Adenocarcinoma/genetics , Animals , Biological Transport , Carbon Radioisotopes , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Fatty Acid Synthase, Type I/antagonists & inhibitors , Fatty Acid Synthase, Type I/genetics , Gene Expression , Gene Expression Profiling , Humans , Male , Mice , Mice, Inbred NOD , Molecular Targeted Therapy , Neoplasms, Experimental , Orlistat , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
9.
Am J Pathol ; 182(1): 172-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23127564

ABSTRACT

C-type natriuretic peptide (CNP) exerts its main biological effects by binding to natriuretic peptide receptor B (NPR-B), a membrane-bound guanylyl cyclase receptor that produces cyclic guanosine monophosphate (cGMP). CNP is known to cause gastrointestinal (GI) smooth muscle relaxation. Experimental evidence suggests a connection between CNP signaling and GI function, with reactive regions in the GI tract possibly affecting transit; however, this relation has not yet been conclusively shown. Here, we show that CNP plays important region-specific roles in the GI tract of mice. We found that treatment with CNP (1 or 2 mg/kg) increased transient cGMP production in the pylorus, colon, and rectum, with the higher dose (2 mg/kg) enhancing gastric emptying in mice; this increase in cGMP levels was however absent in NPR-B-deficient short-limbed dwarfism (SLW) mouse. Furthermore, we found that NPR-B is highly expressed in the pylorus, colon, and rectum, being localized to nerve fibers and to the nuclei and cytoplasm of smooth muscle cells of the GI tract and blood vessels. Our in vivo findings showed that NPR-B-mediated cGMP production after CNP administration specifically acted on the pylorus, colon, and rectum and contributed to gastric emptying. CNP may thus be a potential therapeutic agent for GI motility/transit disorders such as ileus and pyloric stenosis.


Subject(s)
Intestine, Large/physiology , Natriuretic Peptide, C-Type/physiology , Pylorus/physiology , Animals , Cyclic GMP/biosynthesis , Dose-Response Relationship, Drug , Female , Gastric Emptying/drug effects , Gastric Emptying/physiology , Gastrointestinal Agents/administration & dosage , Gastrointestinal Agents/pharmacology , Gastrointestinal Tract/metabolism , Gastrointestinal Transit/drug effects , Gastrointestinal Transit/physiology , Intestine, Large/drug effects , Mice , Mice, Mutant Strains , Natriuretic Peptide, C-Type/administration & dosage , Natriuretic Peptide, C-Type/pharmacology , Pylorus/drug effects , Receptors, Atrial Natriuretic Factor/deficiency , Receptors, Atrial Natriuretic Factor/metabolism , Signal Transduction/physiology
10.
Mol Imaging ; 11(6): 445-50, 2012.
Article in English | MEDLINE | ID: mdl-23084245

ABSTRACT

As cancer stem cells (CSCs) are postulated to play critical roles in cancer development, including metastasis and recurrence, CSC imaging would provide valuable information for cancer treatment and lead to CSC-targeted therapy. To assess the possibility of in vivo CSC targeting, we conducted basic studies on radioimmunotargeting of cancer cells positive for CD133, a CSC marker recognized in various cancers. Antibodies against CD133 were labeled with ¹²5I, and their in vitro cell binding properties were tested. Using the same isotype IgG as a control, in vivo biodistribution of the labeled antibody retaining immunoreactivity was examined in mice bearing an HCT116 xenograft in which a population of the cancer cells expressed CD133. Intratumoral distribution of the labeled antibody was examined and compared to the CD133 expression pattern. The ¹²5I-labeled anti-CD133 antibody showed a modest but significantly higher accumulation in the HCT116 xenograft compared to the control IgG. The intratumoral distribution of the labeled antibody mostly overlapped with the CD133 expression, whereas the control IgG was found in the area close to the necrotic tumor center. Our results indicate that noninvasive in vivo targeting of CSCs could be possible with radiolabeled antibodies against cell membrane markers.


Subject(s)
Antigens, CD/immunology , Glycoproteins/immunology , Immunoconjugates/pharmacology , Neoplastic Stem Cells/radiation effects , Peptides/immunology , AC133 Antigen , Animals , Autoradiography , Cell Line, Tumor , Humans , Immunoconjugates/pharmacokinetics , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/immunology , Tissue Distribution
11.
Nucl Med Commun ; 33(7): 719-25, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22453549

ABSTRACT

OBJECTIVE: The epidermal growth factor receptor (EGFR) is overexpressed in many epithelial cancers, including hepatocellular carcinoma (HCC), and is an attractive target for cancer imaging and therapy. We attempted a novel noninvasive imaging method to evaluate anti-EGFR human monoclonal antibody clones for determining the uptake of therapeutic anti-EGFR antibody in HCC. METHODS: In-vitro cell binding of nine I-labeled antibody clones was compared in the human epidermoid cancer cell line A431, in three HCC cell lines Hep-G2, SK-Hep1, and HuH-7, and in the EGFR-negative control cell line A4. In-labeled or I-labeled 048-006 was subjected to cell binding, competitive inhibition, and internalization assays using A431, SK-Hep1, and HuH-7. Further, In-labeled 048-006 was evaluated in in-vivo biodistribution analysis and single-photon imaging in nude tumor-bearing mice. RESULTS: The 048-006 clone showed the highest binding to EGFR-expressing cells among the nine antibodies. In-labeled or I-labeled 048-006 specifically bound to EGFR-expressing cells with high affinity and was internalized after binding to EGFR. A431 and HuH-7 tumors showed high In-labeled 048-006 uptake, which was visualized by single-photon imaging. CONCLUSION: Radiolabeled human anti-EGFR monoclonal antibody 048-006 has the potential to be a safer imaging probe for predicting tumor uptake of anti-EGFR antibody therapeutic agents in HCC.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Carcinoma, Hepatocellular/diagnostic imaging , Carcinoma, Squamous Cell/diagnostic imaging , ErbB Receptors/metabolism , Liver Neoplasms, Experimental/diagnostic imaging , Tomography, Emission-Computed, Single-Photon/methods , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Humans , Iodine Radioisotopes/pharmacokinetics , Liver Neoplasms, Experimental/metabolism , Mice , Mice, Nude , Tissue Distribution
12.
Anticancer Res ; 31(5): 1549-58, 2011 May.
Article in English | MEDLINE | ID: mdl-21617209

ABSTRACT

BACKGROUND: Dihydroartemisinin (DHA) inhibits the growth of certain cancer cells and xenograft tumors. Further understanding of the molecular mechanisms and genetic participants that govern the antineoplastic effects of DHA is necessary. The anticancer effects of DHA and its underlying mechanisms in pancreatic cancer and the efficacy in animal models by noninvasive optical imaging were evaluated. MATERIALS AND METHODS: Combined with cell/tumor growth assays, flow cytometric analysis, and Hoechst staining, the effect of DHA was investigated using the pancreatic cancer cell line BxPc3-RFP stably expressing red fluorescence protein and in vitro/in vivo optical imaging. Proteins that regulate proliferation (PCNA), apoptosis (Bax and Bcl-2), and angiogenesis (vascular endothelial growth factor (VEGF)) were evaluated in cell and tumor samples by Western blotting and immunohistochemical analyses. RESULTS: DHA inhibited the proliferation and viability of cells in a dose-dependent manner and induced apoptosis. We observed down-regulation of PCNA and Bcl-2, and up-regulation of Bax. VEGF was down-regulated by DHA in cells under normoxic, but not hypoxic, conditions. Fluorescence intensity emitted from cells and tumors correlated linearly with cell count and tumor burden, respectively. CONCLUSION: DHA inhibits cell and tumor growth by interfering with cell proliferation and inducing apoptosis. The antiangiogenic effect of DHA appears to be a complicated process. Optical imaging supports the real-time assessment of DHA efficacy in a preclinical model and comprehensive analysis substantiates that DHA is a potential candidate for pancreatic cancer therapy.


Subject(s)
Antimalarials/therapeutic use , Artemisinins/therapeutic use , Diagnostic Imaging , Disease Models, Animal , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Mice , Mice, Nude , Xenograft Model Antitumor Assays
13.
Nucl Med Biol ; 38(3): 331-7, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21492781

ABSTRACT

INTRODUCTION: C-kit is an important diagnostic and therapeutic target molecule for several malignancies, and c-kit-targeted drugs have been used clinically. Because abundant c-kit expression in tumors is a prerequisite for successful c-kit-targeted therapy, imaging of c-kit expression is expected to play a pivotal role in the therapeutic decision for each patient. We evaluated (64)Cu-labeled Fab of anti-c-kit antibody 12A8 as a positron emission tomography (PET) imaging probe. METHODS: (111)In- or (125)I-Labeled 12A8 Fab was evaluated in vitro by cell binding, competitive inhibition and cellular internalization assays, and in vivo by biodistribution in mice bearing c-kit-expressing and -non-expressing tumors. Next, Fab fragment was labeled with the positron emitter (64)Cu and evaluated by PET. RESULTS: Radiolabeled 12A8 Fab showed specific binding to c-kit-expressing cells with high affinity and internalized into cells after binding to c-kit on cell surface. Although tumor accumulation of [(111)In]Fab was lower than that of [(111)In]IgG, the faster blood clearance of [(111)In]Fab provided higher tumor-to-blood ratio at 6 h postinjection onwards. Blood clearance of (64)Cu-labeled 12A8 Fab was slower than that of [(111)In]Fab, but PET using [(64)Cu]Fab clearly visualized the tumor at 6 h postinjection onwards. CONCLUSION: The (64)Cu-labeled 12A8 Fab could be used for c-kit-specific PET imaging and might help in selecting appropriate patients for c-kit-targeted treatments.


Subject(s)
Antibodies, Monoclonal , Copper Radioisotopes , Gene Expression Regulation, Neoplastic , Immunoglobulin Fab Fragments , Positron-Emission Tomography/methods , Proto-Oncogene Proteins c-kit/immunology , Proto-Oncogene Proteins c-kit/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Cell Line, Tumor , Cell Transformation, Neoplastic , Female , Humans , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fab Fragments/metabolism , Immunoglobulin G/metabolism , Indium Radioisotopes , Iodine Radioisotopes , Mice
14.
Am J Pathol ; 177(2): 822-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20616347

ABSTRACT

Natriuretic peptide receptor B (NPR-B), which has high affinity for C-type natriuretic peptide (CNP) and synthesizes intracellular cGMP, may be involved in gastrointestinal tract (GIT) regulation. A mutant allele of the NPR-B-encoding gene (Npr2) is responsible for the phenotype of the short-limb dwarfism (SLW) mouse. Homozygosity for this autosomal-recessive gene (slw/slw) leads to dwarfism and death before weaning because of milk retention in the stomach and intestinal distention. To elucidate the relationship between CNP/NPR-B signaling and GIT function, we investigated the association between Npr2 mutation and the GIT phenotype in slw/slw mice. The pylorus and large intestine of the mutants did not respond to CNP stimulation; further, they showed pyloric lumen narrowing with randomly aligned circular muscle cells. Comparison of the cGMP and neuronal marker distribution in GIT tissues confirmed cGMP expression in neuronal tissues. An Auerbach's plexus and submucosal tissues of the mutants didn't express cGMP and expressed Ca(2+). In contrast, those of normal mice (controls) expressed both cGMP and Ca(2+). Sequencing revealed that the causative Npr2 mutation was a 7-base deletion in exon 8, resulting in a frameshift and premature termination codon appearance. Therefore, the GIT phenotype of slw/slw mice is because of a CNP/NPR-B-signaling defect caused by an Npr2 mutation. These results facilitate better understanding of the role of CNP/NPR-B signaling in GIT motility.


Subject(s)
Gastrointestinal Diseases/genetics , Mutation , Receptors, Atrial Natriuretic Factor/genetics , Animals , Calcium/metabolism , Cyclic GMP/metabolism , DNA Mutational Analysis , Female , Gastrointestinal Tract/anatomy & histology , Gastrointestinal Tract/physiology , Gastrointestinal Tract/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Muscle, Smooth/physiology , Natriuretic Peptide, C-Type/metabolism , Receptors, Atrial Natriuretic Factor/metabolism
15.
Genomics ; 95(4): 210-6, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20153416

ABSTRACT

Malignant mesothelioma is a highly aggressive tumor arising from serosal surfaces of the pleura and is triggered by past exposure to asbestos. Currently, there is no widely accepted treatment for mesothelioma. Development of effective drug treatments for human cancers requires identification of therapeutic molecular targets. We therefore conducted a large-scale functional screening of mesothelioma cells using a genome-wide small interfering RNA library. We determined that knockdown of 39 genes suppressed mesothelioma cell proliferation. At least seven of the 39 genes-COPA, COPB2, EIF3D, POLR2A, PSMA6, RBM8A, and RPL18A-would be involved in anti-apoptotic function. In particular, the COPA protein was highly expressed in some mesothelioma cell lines but not in a pleural mesothelial cell line. COPA knockdown induced apoptosis and suppressed tumor growth in a mesothelioma mouse model. Therefore, COPA may have the potential of a therapeutic target and a new diagnostic marker of mesothelioma.


Subject(s)
Apoptosis/genetics , Coatomer Protein/genetics , Mesothelioma/genetics , Pleural Neoplasms/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Gene Knockdown Techniques , Humans , Mice , RNA, Small Interfering/genetics
16.
Nucl Med Biol ; 37(2): 179-87, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20152717

ABSTRACT

INTRODUCTION: Gastrointestinal stromal tumor (GIST) is the most common mesenchymal tumor arising from the gastrointestinal tract and highly expresses mutated c-kit. We aimed to develop a specific and sensitive method for detecting GISTs using radiolabeled anti-c-kit monoclonal antibody. METHODS: A mutated c-kit-expressing cell clone was established by transfecting an expressing vector of mutated c-kit gene into HEK293 human embryonic kidney cells. The tumors were developed by inoculating c-kit-expressing cells into nude mice. (125)I- and (111)In-labeled anti-c-kit antibodies (12A8 and 41A11) were evaluated in vitro by cell binding, competitive inhibition and cellular internalization assays, and in vivo by biodistribution and imaging studies in tumor-bearing mice. RESULTS: Both (125)I- and (111)In-labeled antibodies showed specific binding with c-kit-expressing cells with high affinity (dissociation constants = 2.2-7.1x10(9) M(-1)). Internalization assay showed that (125)I-labeled antibodies were rapidly internalized and dehalogenated, with the release of (125)I from the cells, resulting in reduction of cell-associated radioactivity with time. In contrast, (111)In-labeled antibody was internalized but did not result in the reduced radioactivity associated with tumor cells. Reflecting this phenomenon, the in vivo tumor uptake of (125)I-labeled antibody was low on Day 1, further decreasing with time, while tumor uptake of (111)In-labeled antibody was high on Day 1, further increasing with time. The xenografted tumor was clearly visualized by scintigraphy after injection of (111)In-labeled antibody. CONCLUSION: The anti-c-kit monoclonal antibody labeled with a metal radionuclide would be promising for c-kit-targeted imaging of GISTs.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Gastrointestinal Stromal Tumors/metabolism , Indium Radioisotopes/chemistry , Molecular Imaging/methods , Proto-Oncogene Proteins c-kit/immunology , Proto-Oncogene Proteins c-kit/metabolism , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacokinetics , Binding, Competitive , Biological Transport , Cell Line, Tumor , Disease Models, Animal , Gastrointestinal Stromal Tumors/diagnostic imaging , Gastrointestinal Stromal Tumors/genetics , Gastrointestinal Stromal Tumors/pathology , Gene Expression Regulation, Neoplastic , Humans , Iodine Radioisotopes/chemistry , Male , Mice , Mutation , Proto-Oncogene Proteins c-kit/genetics , Tomography, Emission-Computed, Single-Photon
17.
Nucl Med Commun ; 31(5): 380-8, 2010 May.
Article in English | MEDLINE | ID: mdl-20072072

ABSTRACT

BACKGROUND: Malignant mesothelioma is a highly aggressive form of cancer. Curative surgery is the only effective therapy for mesothelioma, and therefore early diagnosis is important. However, early diagnosis is difficult using current diagnostic imaging techniques, and a new imaging method for early diagnosis is urgently required. We evaluated the affinity of radiolabeled monoclonal antibodies to the C-terminal fragment of ERC/mesothelin for this purpose. METHODS: In-labeled or I-labeled IgG against C-terminal fragment of ERC and its Fab fragment were evaluated in vitro by cell binding, competitive inhibition, and cellular internalization assays, and in vivo by biodistribution in mice bearing ERC-expressing tumors. Next, the Fab fragment was labeled with the positron emitter Cu and evaluated by positron emission tomography (PET). RESULTS: Radiolabeled IgG and Fab showed specific binding to ERC-expressing mesothelioma cells with high affinity. Both radiolabeled IgG and Fab internalized into cells after binding to ERC on the cell surface. In-labeled IgG accumulated in ERC-expressing tumors and resulted in a moderate tumor-to-blood ratio at 4 days after injection. Furthermore, PET using Cu-labeled Fab visualized the tumor at 6 h after injection. CONCLUSION: Cu-labeled Fab can be useful for ERC-specific PET imaging, and can thus facilitate improved diagnosis of patients with early-stage mesothelioma.


Subject(s)
Copper Radioisotopes , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/immunology , Membrane Glycoproteins/metabolism , Mesothelioma/diagnostic imaging , Positron-Emission Tomography/methods , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , Disease Models, Animal , Female , GPI-Linked Proteins , Gene Expression Regulation, Neoplastic , Heterocyclic Compounds, 1-Ring/chemistry , Humans , Immunoglobulin Fab Fragments/metabolism , Immunoglobulin G/chemistry , Immunoglobulin G/immunology , Indium Radioisotopes , Iodine Radioisotopes , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/immunology , Mesothelin , Mesothelioma/genetics , Mesothelioma/pathology , Mice , Mice, Inbred BALB C
18.
Nucl Med Biol ; 36(4): 379-88, 2009 May.
Article in English | MEDLINE | ID: mdl-19423005

ABSTRACT

INTRODUCTION: Malignant mesothelioma is a highly aggressive tumor originating in the pleura, peritoneum and pericardium, and the prognosis of patients undergoing current treatment remains poor. To develop new therapies, it is important to have a noninvasive imaging system for evaluating the efficacy of such prospective treatments. We have established clinically relevant mouse models and evaluated conventional and novel positron emission tomography (PET) tracers. METHODS: Epithelioid and sarcomatoid mesothelioma cells were inoculated subcutaneously and intrapleurally into nude mice. Biodistribution and PET imaging studies were conducted by injecting [(18)F]fluoro-2-deoxy-D-glucose (FDG), 3'-[(18)F]fluoro-3'-doxythymidine (FLT) or 4'-methyl-[(11)C]thiothymidine (S-dThd) into the mouse models. In vitro cellular uptake of [(14)C]FDG and [(3)H]FLT and thymidine kinase 1 (TK(1)) activity in both cell lines were measured. Expression of glucose transporter 1 (GLUT-1) and Ki-67 in xenografted tumors was evaluated by immunohistochemical staining. RESULTS: In epithelioid mesothelioma models, biodistribution experiments showed that tumor uptake of [(11)C]S-dThd was significantly higher than that of [(18)F]FDG. On the other hand, in sarcomatoid models, [(18)F]FDG showed significantly higher accumulation than the other two tracers. These differential uptakes of the three tracers were confirmed by PET imaging. The cellular uptake of [(14)C]FDG and [(3)H]FLT and TK(1) activity in sarcomatoid cells were higher than those of epithelioid cells. GLUT-1 protein was strongly expressed in sarcomatoid but not in epithelioid tumor. We observed a high percentage of Ki-67-positive cells in both epithelioid and sarcomatoid tumors. CONCLUSIONS: We established nude mouse models of epithelioid and sarcomatoid subtypes of mesothelioma. PET tracers applicable for the evaluation of epithelioid and sarcomatoid mesothelioma would vary: [(18)F]FLT and [(11)C]S-dThd seemed suitable for the epithelioid subtype and [(18)F]FDG seemed suitable for the sarcomatoid subtype in our mouse models. Our results indicated that cellular uptake and TK(1) activity in vitro are not always consistent with tracer uptake of [(18)F]FLT and [(11)C]S-dThd in vivo. These mouse models and PET imaging might be useful tools for evaluating new and effective treatments in mesothelioma.


Subject(s)
Mesothelioma/diagnostic imaging , Positron-Emission Tomography/methods , Animals , Cell Line, Tumor , GPI-Linked Proteins , Gene Expression Regulation, Neoplastic , Glucose Transporter Type 1/immunology , Humans , Injections, Subcutaneous , Ki-67 Antigen/immunology , Male , Membrane Glycoproteins/metabolism , Mesothelin , Mesothelioma/genetics , Mesothelioma/pathology , Mesothelioma/surgery , Mice , Mice, Nude , Pleural Cavity , Radioactive Tracers , Transplantation, Heterologous
19.
J Nucl Med ; 50(5): 827-30, 2009 May.
Article in English | MEDLINE | ID: mdl-19372488

ABSTRACT

UNLABELLED: Acute allograft rejection remains a major complication after liver transplantation. We report a semiquantitative imaging method of detecting acute allograft rejection with (18)F-FDG PET. METHODS: Syngeneic and allogeneic transplanted rats, with or without immunosuppressive treatment, were subjected to serial PET. Autoradiography of the liver was conducted in both the syngeneic and the allogeneic rats. RESULTS: A significant increment of (18)F-FDG accumulation in liver allografts was observed by PET on day 2. The (18)F-FDG signal was concentrated in the area where inflammatory cells around the vessels were detected by autoradiography. Allotransplanted rats treated with an immunosuppressive agent displayed a marked decrease in hepatic (18)F-FDG uptake, compared with allotransplanted rats that were not treated. CONCLUSION: (18)F-FDG PET may be a valid method for facilitating the development of protocols to diagnose graft rejection and to monitor the efficacy of immunosuppressive therapy.


Subject(s)
Fluorodeoxyglucose F18 , Graft Rejection/diagnostic imaging , Graft Rejection/prevention & control , Immunosuppressive Agents/administration & dosage , Liver Transplantation/adverse effects , Liver Transplantation/diagnostic imaging , Positron-Emission Tomography/methods , Animals , Disease Models, Animal , Graft Rejection/etiology , Humans , Image Interpretation, Computer-Assisted/methods , Male , Prognosis , Radiopharmaceuticals , Rats , Rats, Inbred Lew , Treatment Outcome
20.
J Hered ; 98(6): 575-80, 2007.
Article in English | MEDLINE | ID: mdl-17728275

ABSTRACT

Short-limbed dwarfism (SLW) is a new mutant mouse characterized by a dwarf phenotype with markedly short body, limbs, and tail. In the present study, we investigated the skeletal phenotypes of the SLW mouse and determined the chromosomal localization to identify the gene responsible for the phenotypes (slw). Skeletal preparations stained with alcian blue and alizarin red revealed that longitudinal growth of the extremities of the affected (slw/slw) mice was significantly reduced in comparison with that of normal mice, whereas the positions and numbers of skeletal elements were normal. Histological examination of tibial growth plates of the affected mice showed that the numbers of proliferating and hypertrophic chondrocytes were obviously diminished. These phenotypes resembled to those of human chondrodysplasias caused by defective chondrocyte proliferation and differentiation. We mapped the slw locus on an 11.7-cM interval of the proximal region of mouse chromosome 4 by linkage analysis. Furthermore, allelism test using Npr2(cn) locus, a mutant allele of Npr2 gene encoding a natriuretic peptide receptor B, revealed that slw locus is an allele of the Npr2 gene. These results suggest that the dwarf phenotype of the SLW mouse is caused by the disturbed endochondral ossification, and a mutation in the Npr2 gene is expected to be responsible for the phenotypes of the SLW mouse.


Subject(s)
Dwarfism/genetics , Exostoses, Multiple Hereditary/genetics , Guanylate Cyclase/genetics , Receptors, Atrial Natriuretic Factor/genetics , Animals , Chromosome Mapping , Crosses, Genetic , Exostoses, Multiple Hereditary/pathology , Extremities/anatomy & histology , Female , Growth Plate/pathology , Male , Mice , Mice, Mutant Strains/anatomy & histology , Mice, Mutant Strains/genetics , Probability
SELECTION OF CITATIONS
SEARCH DETAIL
...