Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Transl Med ; 17(1): 425, 2019 12 19.
Article in English | MEDLINE | ID: mdl-31878948

ABSTRACT

BACKGROUND: Spontaneously metastatic xenograft models of cancer are infrequent and the few that exist are resource intensive. In xenografts, caliper measurements can be used to determine primary tumor burden and response to therapy but in metastatic disease models determination of the presence of metastatic disease, metastatic burden, and response to therapy are difficult, often requiring serial necropsy. In this study we characterized the development of visceral metastases in a patient derived xenograft model (PDXM) using in vivo imaging. RESULTS: We identified and characterized the previously unreported development of spontaneous liver and bone metastasis in a known patient derived xenograft, bladder xenograft BL0293F, developed by Jackson Laboratories and the University of California at Davis and available from the National Cancer Institute Patient-Derived Models Repository [1]. Among FDG-PET/CT, contrast-enhanced MRI and non-contrast MRI, non-contrast T2w MRI was the most effective and efficient imaging technique. On non-contrast T2 weighted MRI, hepatic metastases were observed in over 70% of animals at 52 days post tumor implantation without resection of the xenograft and in 100% of animals at day 52 following resection of the xenograft. In a group of animals receiving one cycle of effective chemotherapy, no animals demonstrated metastasis by imaging, confirming the utility of this model for therapy evaluation. There was good agreement between pathologic grade and extent of involvement observed on MRI T2w imaging. CONCLUSION: PDX BL0293F is a reliable visceral organ (liver) metastatic model with high penetrance in both non-aggravated and post excisional situations, providing a reliable window for therapy intervention prior to required excision of the xenograft. The imaging characteristics of this model are highly favorable for non-clinical research studies of metastatic disease when used in conjunction with non-contrast T2 weighted MRI.


Subject(s)
Imaging, Three-Dimensional , Urinary Bladder Neoplasms/diagnostic imaging , Urinary Bladder Neoplasms/pathology , Animals , Female , Humans , Liver Neoplasms/secondary , Magnetic Resonance Imaging , Mice, Inbred NOD , Neoplasm Metastasis , Xenograft Model Antitumor Assays
2.
Mol Cancer Res ; 17(8): 1613-1626, 2019 08.
Article in English | MEDLINE | ID: mdl-31043488

ABSTRACT

Renal cell carcinoma (RCC) associated with Xp11.2 translocation (TFE3-RCC) has been recently defined as a distinct subset of RCC classified by characteristic morphology and clinical presentation. The Xp11 translocations involve the TFE3 transcription factor and produce chimeric TFE3 proteins retaining the basic helix-loop-helix leucine zipper structure for dimerization and DNA binding suggesting that chimeric TFE3 proteins function as oncogenic transcription factors. Diagnostic biomarkers and effective forms of therapy for advanced cases of TFE3-RCC are as yet unavailable. To facilitate the development of molecular based diagnostic tools and targeted therapies for this aggressive kidney cancer, we generated a translocation RCC mouse model, in which the PRCC-TFE3 transgene is expressed specifically in kidneys leading to the development of RCC with characteristic histology. Expression of the receptor tyrosine kinase Ret was elevated in the kidneys of the TFE3-RCC mice, and treatment with RET inhibitor, vandetanib, significantly suppressed RCC growth. Moreover, we found that Gpnmb (Glycoprotein nonmetastatic B) expression was notably elevated in the TFE3-RCC mouse kidneys as seen in human TFE3-RCC tumors, and confirmed that GPNMB is the direct transcriptional target of TFE3 fusions. While GPNMB IHC staining was positive in 9/9 cases of TFE3-RCC, Cathepsin K, a conventional marker for TFE3-RCC, was positive in only 67% of cases. These data support RET as a potential target and GPNMB as a diagnostic marker for TFE3-RCC. The TFE3-RCC mouse provides a preclinical in vivo model for the development of new biomarkers and targeted therapeutics for patients affected with this aggressive form of RCC. IMPLICATIONS: Key findings from studies with this preclinical mouse model of TFE3-RCC underscore the potential for RET as a therapeutic target for treatment of patients with TFE3-RCC, and suggest that GPNMB may serve as diagnostic biomarker for TFE3 fusion RCC.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Carcinoma, Renal Cell/pathology , Chromosomes, Human, X , Kidney Neoplasms/pathology , Membrane Glycoproteins/metabolism , Oncogene Proteins, Fusion , Translocation, Genetic , Adolescent , Adult , Aged , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Cell Cycle Proteins/genetics , Cell Proliferation , Child , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Male , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Middle Aged , Neoplasm Proteins/genetics , Prognosis , Survival Rate , Tumor Cells, Cultured , Young Adult
3.
J Clin Invest ; 128(9): 3794-3805, 2018 08 31.
Article in English | MEDLINE | ID: mdl-29920191

ABSTRACT

Control of cellular metabolism is critical for efficient cell function, although little is known about the interplay between cell subset-specific metabolites in situ, especially in the tumor setting. Here, we determined how a macrophage-specific (Mϕ-specific) metabolite, itaconic acid, can regulate tumor progression in the peritoneum. We show that peritoneal tumors (B16 melanoma or ID8 ovarian carcinoma) elicited a fatty acid oxidation-mediated increase in oxidative phosphorylation (OXPHOS) and glycolysis in peritoneal tissue-resident macrophages (pResMϕ). Unbiased metabolomics identified itaconic acid, the product of immune-responsive gene 1-mediated (Irg1-mediated) catabolism of mitochondrial cis-aconitate, among the most highly upregulated metabolites in pResMϕ of tumor-bearing mice. Administration of lentivirally encoded Irg1 shRNA significantly reduced peritoneal tumors. This resulted in reductions in OXPHOS and OXPHOS-driven production of ROS in pResMϕ and ROS-mediated MAPK activation in tumor cells. Our findings demonstrate that tumors profoundly alter pResMϕ metabolism, leading to the production of itaconic acid, which potentiates tumor growth. Monocytes isolated from ovarian carcinoma patients' ascites fluid expressed significantly elevated levels of IRG1. Therefore, IRG1 in pResMϕ represents a potential therapeutic target for peritoneal tumors.


Subject(s)
Macrophages, Peritoneal/metabolism , Peritoneal Neoplasms/metabolism , Succinates/metabolism , Animals , Carboxy-Lyases , Cell Line, Tumor , Disease Progression , Fatty Acids/metabolism , Female , Glycolysis , Humans , Hydro-Lyases/deficiency , Hydro-Lyases/genetics , Hydro-Lyases/metabolism , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Ovarian Neoplasms/metabolism , Oxidative Phosphorylation , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/pathology , Proteins/metabolism , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Tumor Burden
4.
Nat Protoc ; 9(11): 2682-92, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25340441

ABSTRACT

We have developed a reliable noninvasive method for monitoring colonic tumors and mucosal inflammation in a mouse model of colon cancer using magnetic resonance colonography (MRC). After a mild cleansing enema, the colon is filled with Fluorinert, a perfluorinated liquid that does not produce a proton MR signal. The mouse is placed in a dedicated volume MR receiver coil, and high-resolution images are acquired in three planes. The Fluorinert enema distends the mouse colon, creating an artifact-free black homogeneous background, allowing clear delineation of the inflamed colonic wall and visualization of luminal tumors in various stages of development. A gadolinium-based contrast agent can be administered i.v. to the animal to detect mural inflammation or tumor vascularity. This technique is useful for serial monitoring of the effects of preventive or therapeutic strategies on tumor development without killing the animal or requiring invasive endoscopies. The animal preparation and imaging can be completed in ∼1.5 h.


Subject(s)
Colonic Neoplasms/diagnosis , Colonography, Computed Tomographic/methods , Magnetic Resonance Imaging/methods , Animals , Colonic Neoplasms/pathology , Disease Models, Animal , Enema , Equipment Design , Female , Fluorocarbons , Magnetic Resonance Imaging/instrumentation , Male , Mice, Inbred Strains , Time Factors
5.
Int J Mol Imaging ; 2014: 102702, 2014.
Article in English | MEDLINE | ID: mdl-24724022

ABSTRACT

Metastatic spread is the leading cause of death from cancer. Early detection of cancer at primary and metastatic sites by noninvasive imaging modalities would be beneficial for both therapeutic intervention and disease management. Noninvasive imaging modalities such as bioluminescence (optical), positron emission tomography (PET)/X-ray computed tomography (CT), and magnetic resonance imaging (MRI) can provide complementary information and accurately measure tumor growth as confirmed by histopathology. Methods. We validated two metastatic tumor models, MDA-MD-231-Luc and B16-F10-Luc intravenously injected, and 4T1-Luc cells orthotopically implanted into the mammary fat pad. Longitudinal whole body bioluminescence imaging (BLI) evaluated metastasis, and tumor burden of the melanoma cell line (B16-F10-Luc) was correlated with (PET)/CT and MRI. In addition, ex vivo imaging evaluated metastasis in relevant organs and histopathological analysis was used to confirm imaging. Results. BLI revealed successful colonization of cancer cells in both metastatic tumor models over a 4-week period. Furthermore, lung metastasis of B16-F10-Luc cells imaged by PET/CT at week four showed a strong correlation (R (2) = 0.9) with histopathology. The presence and degree of metastasis as determined by imaging correlated (R (2) = 0.7) well with histopathology findings. Conclusions. We validated two metastatic tumor models by longitudinal noninvasive imaging with good histopathology correlation.

6.
Cancer Res ; 73(17): 5473-84, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23824743

ABSTRACT

Analysis of the Polyp Prevention Trial showed an association between an isorhamnetin-rich diet and a reduced risk of advanced adenoma recurrence; however, the mechanism behind the chemoprotective effects of isorhamnetin remains unclear. Here, we show that isorhamnetin prevents colorectal tumorigenesis of FVB/N mice treated with the chemical carcinogen azoxymethane and subsequently exposed to colonic irritant dextran sodium sulfate (DSS). Dietary isorhamnetin decreased mortality, tumor number, and tumor burden by 62%, 35%, and 59%, respectively. MRI, histopathology, and immunohistochemical analysis revealed that dietary isorhamnetin resolved the DSS-induced inflammatory response faster than the control diet. Isorhamnetin inhibited AOM/DSS-induced oncogenic c-Src activation and ß-catenin nuclear translocation, while promoting the expression of C-terminal Src kinase (CSK), a negative regulator of Src family of tyrosine kinases. Similarly, in HT-29 colon cancer cells, isorhamnetin inhibited oncogenic Src activity and ß-catenin nuclear translocation by inducing expression of csk, as verified by RNA interference knockdown of csk. Our observations suggest the chemoprotective effects of isorhamnetin in colon cancer are linked to its anti-inflammatory activities and its inhibition of oncogenic Src activity and consequential loss of nuclear ß-catenin, activities that are dependent on CSK expression.


Subject(s)
Colorectal Neoplasms/prevention & control , Phytotherapy , Plant Extracts/pharmacology , Quercetin/analogs & derivatives , beta Catenin/metabolism , src-Family Kinases/metabolism , Animals , Apoptosis/drug effects , Azoxymethane/toxicity , Blotting, Western , CSK Tyrosine-Protein Kinase , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/metabolism , Dextran Sulfate , Humans , Immunoenzyme Techniques , Male , Mice , Protein Transport , Quercetin/pharmacology , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , beta Catenin/genetics , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics
7.
Invest Radiol ; 48(11): 745-54, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23748228

ABSTRACT

OBJECTIVE: Macromolecular contrast agents for magnetic resonance imaging (MRI) are useful blood-pool agents because of their long systemic half-life and have found applications in monitoring tumor vasculature and angiogenesis. Macromolecular contrast agents have been able to overcome some of the disadvantages of the conventional small-molecule contrast agent Magnevist (gadolinium-diethylenetriaminepentaacetic acid), such as rapid extravasation and quick renal clearance, which limits the viable MRI time. There is an urgent need for new MRI contrast agents that increase the sensitivity of detection with a higher relaxivity, longer blood half-life, and reduced toxicity from free Gd3+ ions. Here, we report on the characterization of a novel water-soluble, derivatized, gadolinium-enclosed metallofullerene nanoparticle (Hydrochalarone-1) in development as an MRI contrast agent. MATERIALS AND METHODS: The physicochemical properties of Hydrochalarone-1 were characterized by dynamic light scattering (hydrodynamic diameter), atomic force microscopy (particle height), ζ potential analysis (surface charge), and inductively coupled plasma-mass spectrometry (gadolinium concentration). The blood compatibility of Hydrochalarone-1 was also assessed in vitro through analysis of hemolysis, platelet aggregation, and complement activation of human blood. In vitro relaxivities, in vivo pharmacokinetics, and a pilot in vivo acute toxicity study were also performed. RESULTS: An extensive in vitro and in vivo characterization of Hydrochalarone-1 is described here. The hydrodynamic size of Hydrochalarone-1 was 5 to 7 nm depending on the dispersing media, and it was negatively charged at physiological pH. Hydrochalarone-1 showed compatibility with blood cells in vitro, and no significant hemolysis, platelet aggregation, or complement activation was observed in vitro. In addition, Hydrochalarone-1 had significantly higher r1 and r2 in vitro relaxivities in human plasma in comparison with Magnevist and was not toxic at the doses administered in an in vivo pilot acute-dose toxicity study in mice.In vivo MRI pharmacokinetic analysis after a single intravenous injection of Hydrochalarone-1 (0.2 mmol Gd/kg) showed that the volume of distribution at steady state was approximately 100 mL/kg, suggesting prolonged systemic circulation. Hydrochalarone-1 also had a long blood half-life (88 minutes) and increased relaxivity, suggesting application as a promising blood-pool MRI contrast agent. CONCLUSIONS: The evidence suggests that Hydrochalarone-1, with its long systemic half-life, may have significant utility as a blood-pool MRI contrast agent.


Subject(s)
Contrast Media/chemistry , Fullerenes/chemistry , Gadolinium/chemistry , Magnetic Resonance Imaging , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacokinetics , Animals , Complement Activation , Contrast Media/pharmacokinetics , Contrast Media/toxicity , Fullerenes/pharmacokinetics , Fullerenes/toxicity , Gadolinium/pharmacokinetics , Gadolinium/toxicity , Hemolysis , Humans , Macromolecular Substances , Mice , Microscopy, Atomic Force , Molecular Dynamics Simulation , Nanoparticles , Organometallic Compounds/toxicity , Platelet Aggregation , Spectrophotometry, Atomic
8.
PLoS One ; 6(10): e25625, 2011.
Article in English | MEDLINE | ID: mdl-21998674

ABSTRACT

Pazopanib is an FDA approved Vascular Endothelial Growth Factor Receptor inhibitor. We previously reported that it also inhibits tumor cell B-Raf activity in an experimental brain metastatic setting. Here, we determine the effects of different B-Raf genotypes on pazopanib efficacy, in terms of primary tumor growth and anti-angiogenesis. A panel of seven human breast cancer and melanoma cell lines harboring different mutations in the Ras-Raf pathway was implanted orthotopically in mice, and tumor growth, ERK1/2, MEK1/2 and AKT activation, and blood vessel density and permeability were analyzed. Pazopanib was significantly inhibitory to xenografts expressing either exon 11 mutations of B-Raf, or HER2 activated wild type B-Raf; no significant inhibition of a xenograft expressing the common V600E B-Raf mutation was observed. Decreased pMEK staining in the responsive tumors confirmed that B-Raf was targeted by pazopanib. Interestingly, pazopanib inhibition of tumor cell B-Raf also correlated with its anti-angiogenic activity, as quantified by vessel density and area. In conclusion, using pazopanib, tumor B-Raf status was identified as a significant determinant of both tumor growth and angiogenesis.


Subject(s)
Breast Neoplasms/pathology , Melanoma/pathology , Neovascularization, Pathologic/drug therapy , Proto-Oncogene Proteins B-raf/metabolism , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Xenograft Model Antitumor Assays , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/blood supply , Breast Neoplasms/enzymology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Indazoles , MAP Kinase Signaling System/drug effects , Melanoma/blood supply , Melanoma/enzymology , Mice , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Pyrimidines/therapeutic use , Sulfonamides/therapeutic use
9.
Clin Cancer Res ; 17(1): 142-53, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21081656

ABSTRACT

PURPOSE: Brain metastases of breast cancer contribute significantly to patient morbidity and mortality. We have tested pazopanib, a recently approved antiangiogenic drug that targets VEGFR1, VEGFR2, VEGFR3, PDGFRß, PDGFRα, and c-kit, for prevention of experimental brain metastases and mechanism of action. EXPERIMENTAL DESIGN: In vitro assays included B-Raf enzymatic assays, Western blots, and angiogenesis assays. For in vivo assays, HER2 transfectants of the brain seeking sublines of MDA-MB-231 cells (231-BR-HER2) and MCF7 cells (MCF7-HER2-BR3, derived herein) were injected into the left cardiac ventricle of mice and treated with vehicle or pazopanib beginning on day 3 postinjection. Brain metastases were counted histologically, imaged, and immunostained. RESULTS: Treatment with 100 mg/kg of pazopanib resulted in a 73% decline in large 231-BR-HER2 metastases (P < 0.0001) and a 39% decline in micrometastases (P = 0.004). In vitro, pazopanib was directly antiproliferative to 231-BR-HER2 breast cancer cells and inhibited MEK and ERK activation in vitro despite B-Raf and Ras mutations. Enzymatic assays demonstrated that pazopanib directly inhibited the wild type and exon 11 oncogenic mutant, but not the V600E mutant forms of B-Raf. Activation of the B-Raf targets pERK1/2 and pMEK1/2 was decreased in pazopanib-treated brain metastases whereas blood vessel density was unaltered. In the MCF7-HER2-BR3 experimental brain metastasis model, pazopanib reduced overall brain metastasis volume upon magnetic resonance imaging (MRI) by 55% (P = 0.067), without affecting brain metastasis vascular density. CONCLUSIONS: The data identify a new activity for pazopanib directly on tumor cells as a pan-Raf inhibitor and suggest its potential for prevention of brain metastatic colonization of HER2(+) breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/prevention & control , Brain Neoplasms/secondary , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Pyrimidines/pharmacology , Receptor, ErbB-2/genetics , Sulfonamides/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Brain Neoplasms/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Endothelial Cells/drug effects , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Indazoles , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation , Proto-Oncogene Proteins B-raf/metabolism , Pyrimidines/therapeutic use , Signal Transduction/drug effects , Sulfonamides/therapeutic use , Xenograft Model Antitumor Assays
10.
Bioconjug Chem ; 20(7): 1412-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19555072

ABSTRACT

In this paper, we report a new method to prepare and characterize a contrast agent based on a fourth-generation (G4) polyamidoamine (PAMAM) dendrimer conjugated to the gadolinium complex of the bifunctional diethylenetriamine pentaacetic acid derivative (1B4M-DTPA). The method involves preforming the metal-ligand chelate in alcohol prior to conjugation to the dendrimer. The dendrimer-based agent was purified by a Sephadex G-25 column and characterized by elemental analysis. The analysis and SE-HPLC data gave a chelate to dendrimer ratio of 30:1 suggesting conjugation at approximately every other amine terminal on the dendrimer. Molar relaxivity of the agent measured at pH 7.4 displayed a higher value than that of the analogous G4 dendrimer based agent prepared by the postmetal incorporation method (r(1) = 26.9 vs 13.9 mM(-1) s(-1) at 3 T and 22 degrees C). This is hypothesized to be due to the higher hydrophobicity of this conjugate and the lack of available charged carboxylate groups from noncomplexed free ligands that might coordinate to the metal and thus also reduce water exchange sites. Additionally, the distribution populations of compounds that result from the postmetal incorporation route are eliminated from the current product simplifying characterization as quality control issues pertaining to the production of such agents for clinical use as MR contrast agents. In vivo imaging in mice showed a reasonably fast clearance (t(1/2) = 24 min) suggesting a viable agent for use in clinical application.


Subject(s)
Contrast Media/chemical synthesis , Contrast Media/pharmacokinetics , Pentetic Acid/analogs & derivatives , Polyamines/chemistry , Animals , Contrast Media/chemistry , Dendrimers , Gadolinium/chemistry , Gadolinium/pharmacokinetics , Magnetic Resonance Imaging , Mice , Pentetic Acid/chemical synthesis , Pentetic Acid/chemistry , Pentetic Acid/pharmacokinetics , Polyamines/chemical synthesis , Polyamines/pharmacokinetics , Whole Body Imaging
11.
Neoplasia ; 11(3): 237-46, 1p following 246, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19242605

ABSTRACT

Early detection of precancerous tissue has significantly improved survival of most cancers including colorectal cancer (CRC). Animal models designed to study the early stages of cancer are valuable for identifying molecular events and response indicators that correlate with the onset of disease. The goal of this work was to investigate magnetic resonance (MR) colonography in a mouse model of CRC on a clinical MR imager. Mice treated with azoxymethane and dextran sulfate sodium were imaged by serial MR colonography (MRC) from initiation to euthanasia. Magnetic resonance colonography was obtained with both T1- and T2-weighted images after administration of a Fluorinert enema to remove residual luminal signal and intravenous contrast to enhance the colon wall. Individual tumor volumes were calculated and validated ex vivo. The Fluorinert enema provided a clear differentiation of the lumen of the colon from the mucosal lining. Inflammation was detected 3 days after dextran sulfate sodium exposure and subsided during the next week. Tumors as small as 1.2 mm(3) were detected and as early as 29 days after initiation. Individual tumor growths were followed over time, and tumor volumes were measured by MR imaging correlated with volumes measured ex vivo. The use of a Fluorinert enema during MRC in mice is critical for differentiating mural processes from intraluminal debris. Magnetic resonance colonography with Fluorinert enema and intravenous contrast enhancement will be useful in the study of the initial stages of colon cancer and will reduce the number of animals needed for preclinical trials of prevention or intervention.


Subject(s)
Colitis/diagnosis , Colonic Neoplasms/diagnosis , Early Detection of Cancer , Inflammation/diagnosis , Magnetic Resonance Imaging , Animals , Colitis/complications , Contrast Media/administration & dosage , Disease Models, Animal , Disease Progression , Enema , Fluorocarbons/administration & dosage , Mice
12.
J Natl Cancer Inst ; 100(2): 140-54, 2008 Jan 16.
Article in English | MEDLINE | ID: mdl-18182616

ABSTRACT

BACKGROUND: Patients with Birt-Hogg-Dubé (BHD) syndrome harbor germline mutations in the BHD tumor suppressor gene that are associated with an increased risk for kidney cancer. BHD encodes folliculin, a protein that may interact with the energy- and nutrient-sensing 5'-AMP-activated protein kinase-mammalian target of rapamycin (AMPK-mTOR) signaling pathways. METHODS: We used recombineering methods to generate mice with a conditional BHD allele and introduced the cadherin 16 (KSP)-Cre transgene to target BHD inactivation to the kidney. Kidney cell proliferation was measured by BrdU incorporation and phospho-histone H3 staining. Kidney weight data were analyzed with Wilcoxon's rank-sum, Student's t, and Welch's t tests. Hematoxylin and eosin staining and immunoblot analysis and immunohistochemistry of cell cycle and signaling proteins were performed on mouse kidney cells and tissues. BHD knockout mice and kidney cells isolated from BHD knockout and control mice were treated with the mTOR inhibitor rapamycin. Mouse survival was evaluated by Kaplan-Meier analyses. All statistical tests were two-sided. RESULTS: BHD knockout mice developed enlarged polycystic kidneys and died from renal failure by 3 weeks of age. Targeted BHD knockout led to the activation of Raf-extracellular signal-regulated protein kinase (Erk)1/2 and Akt-mTOR pathways in the kidneys and increased expression of cell cycle proteins and cell proliferation. Rapamycin-treated BHD knockout mice had smaller kidneys than buffer-treated BHD knockout mice had (n = 4-6 mice per group, relative kidney/body weight ratios, mean = 4.64% vs 12.2%, difference = 7.6%, 95% confidence interval = 5.2% to 10.0%; P < .001) and longer median survival time (n = 4-5 mice per group, 41.5 vs 23 days; P = .0065 ). CONCLUSIONS: Homozygous loss of BHD may initiate renal tumorigenesis in the mouse. The conditional BHD knockout mouse may be a useful research model for dissecting multistep kidney carcinogenesis, and rapamycin may be considered as a potential treatment for Birt-Hogg-Dubé syndrome.


Subject(s)
Cell Proliferation , Gene Silencing , Kidney Neoplasms/genetics , Kidney/metabolism , Kidney/pathology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Polycystic Kidney Diseases/genetics , Protein Kinases/metabolism , Proto-Oncogene Proteins/genetics , Tumor Suppressor Proteins/genetics , Animals , Antibiotics, Antineoplastic/pharmacology , Blotting, Southern , Cell Proliferation/drug effects , Disease Models, Animal , Estrone/genetics , Fluorescent Antibody Technique , Genotype , Germ-Line Mutation , Immunoblotting , Immunohistochemistry , Kaplan-Meier Estimate , Kidney/drug effects , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 3/drug effects , Polycystic Kidney Diseases/complications , Polycystic Kidney Diseases/metabolism , Polycystic Kidney Diseases/pathology , Protein Kinases/drug effects , Random Allocation , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Sirolimus/pharmacology , Syndrome , TOR Serine-Threonine Kinases
13.
Mol Imaging ; 5(1): 41-52, 2006.
Article in English | MEDLINE | ID: mdl-16779969

ABSTRACT

The development of improvements in magnetic resonance imaging (MRI) that would enhance sensitivity, leading to earlier detection of cancer and visualization of metastatic disease, is an area of intense exploration. We have devised a tumor-targeting, liposomal nanodelivery platform for use in gene medicine. This systemically administered nanocomplex has been shown to specifically and efficiently deliver both genes and oligonucleotides to primary and metastatic tumor cells, resulting in significant tumor growth inhibition and even tumor regression. Here we examine the effect on MRI of incorporating conventional MRI contrast agent Magnevist into our anti-transferrin receptor single-chain antibody (TfRscFv) liposomal complex. Both in vitro and in an in vivo orthotopic mouse model of pancreatic cancer, we show increased resolution and image intensity with the complexed Magnevist. Using advanced microscopy techniques (scanning electron microscopy and scanning probe microscopy), we also established that the Magnevist is in fact encapsulated by the liposome in the complex and that the complex still retains its nanodimensional size. These results demonstrate that this TfRscFv-liposome-Magnevist nanocomplex has the potential to become a useful tool in early cancer detection.


Subject(s)
Drug Delivery Systems/methods , Magnetic Resonance Imaging/methods , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/therapy , Animals , Cell Line, Tumor , Early Diagnosis , Gadolinium DTPA , Genetic Therapy/methods , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/immunology , K562 Cells , Liposomes/chemistry , Liver Neoplasms/diagnosis , Liver Neoplasms/secondary , Mice , Mice, Nude , Microscopy, Electron, Scanning , Microscopy, Scanning Probe , Nanotechnology/methods , Pancreatic Neoplasms/genetics , Receptors, Transferrin/genetics , Receptors, Transferrin/immunology , Xenograft Model Antitumor Assays
14.
Prostate ; 66(7): 708-17, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16425198

ABSTRACT

BACKGROUND: Mouse prostate cancer modeling presents unique obstacles to the study of spontaneous tumor initiation and progression due to the anatomical location of the tissue. RESULTS: High resolution (130 microm(x) x 130 microm(y) x 300 microm(z)), three-dimensional MRI allowed for the visualization, segmentation, and volumetric measurement of the prostate from normal and genetically engineered animals, in vivo. Additionally, MRS performed on the prostate epithelia of probasin-ErbB-2Delta x Pten(+/-) mice identified changes in the relative concentrations of the metabolites choline and citrate, which was not observed in TRAMP mice. METHODS: T1-weighted MRI was performed on normal, TRAMP, probasin-ErbB-2/Her2/Neu (probasin-ErbB-2Delta), and probasin-ErbB-2Delta in the context of decreased Pten activity (probasin-ErbB-2Delta x Pten(+/-)) mice. Volume-localized single-voxel proton magnetic resonance spectroscopy (SVS (1)H MRS) was also performed. CONCLUSIONS: The data presented supports the use of combined MRI and MRS for the measurement of biochemical and morphometric alterations in mouse models of prostate cancer.


Subject(s)
Disease Models, Animal , Prostatic Neoplasms/pathology , Prostatic Neoplasms/veterinary , Animals , Imaging, Three-Dimensional , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Male , Mice
15.
Cell Cycle ; 5(1): 113-9, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16340310

ABSTRACT

The development of effective cancer therapies has been hampered, in part, by the inability to noninvasively follow tumor progression from the initial cancerous lesion through to metastasis. We have previously shown that superparamagnetic iron oxide particles can be used as magnetic resonance imaging contrast agents to label embryonic, mesenchymal and hematopoietic stem cells in vivo. Improving the capacity to non-invasively image cancer progression is an appealing method that could be useful for assessing the efficacy of anticancer therapies. We have established that human prostate (LNCaP, DU145, PC3), rodent prostate (TRAMPC1, YPEN-1), human breast (MDA-MB-231) and mouse mammary (Myc/VEGF) cancer cell lines were readily labeled by fluorescent superparamagnetic sub-micron particles of iron oxide (MPIOs). The MPIOs were essentially inert with respect to cell proliferation and tumor formation. Fluorescence stereomicroscopy and three dimensional magnetic resonance imaging (MRI) determined that subcutaneous, intramuscular or orthotopically implanted labeled cancer cells could be imaged, in vivo, despite in some cases being undetectable by manual palpation. The MPIO-labeled cancer cells could also be imaged, in vivo, at least 6 weeks after implantation. The fluorescent MPIOs further allowed for the ex vivo identification of tumors cells from histological sections. This study demonstrates the feasibility of using fluorescent MPIOs in prostate and breast cancer cell lines as both a negative contrast agent for in vivo MRI as well as a fluorescent tumor marker for optical imaging in vivo and ex vivo.


Subject(s)
Breast Neoplasms/pathology , Contrast Media/analysis , Image Enhancement/methods , Magnetic Resonance Imaging/methods , Prostatic Neoplasms/pathology , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/ultrastructure , Cell Line, Tumor , Estrogens/metabolism , Humans , Male , Mice , Microscopy, Atomic Force , Microscopy, Electron, Transmission , Neoplasm Transplantation , Prostatic Neoplasms/ultrastructure , Proto-Oncogene Proteins c-myc/metabolism , Rats , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...