Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Gynecol Oncol ; 155(2): 331-339, 2019 11.
Article in English | MEDLINE | ID: mdl-31493899

ABSTRACT

INTRODUCTION: PI3K pathway signaling has received attention as a molecular target in clear cell ovarian carcinoma (CCOC). MDM2 is one of the AKT effectors in the PI3K pathway, which binds to and degrades p53. In this study, we aimed to clarify the prognostic significance of PIK3CA and MDM2 expression, and potential therapeutic effect of a dual inhibition of the PI3K pathway and MDM2. MATERIALS AND METHODS: cDNA expression was evaluated by using microarray data using 75 samples of CCOC. DS-7423 (dual inhibitor of pan-PI3K and mTOR) and RG7112 (MDM2 inhibitor) were used on CCOC cell lines to evaluate cell proliferation, expression level of MDM2 related proteins, and apoptosis by MTT assay, western blotting, and flow cytometry. DS-7423 (3 mg/kg) and/or RG7112 (50 mg/kg) were orally administrated every day for three weeks, and the anti-tumor effect was evaluated using tumor xenografts, along with immunohistochemistry. RESULTS: Tumors with high expression of both PIK3CA and MDM2 showed significantly worse prognosis in expression array of 71 CCOCs (P = 0.013). Dual inhibition of the PI3K pathway by DS-7423 and MDM2 by RG7112 showed synergistic anti-proliferative effect in 4 CCOC cell lines without TP53 mutations. The combination therapy more robustly induced pro-apoptotic proteins (PUMA and cleaved PARP) with increase of sub G1 population and apoptotic cells, compared with either single agent alone. The combination therapy significantly reduced tumor volume in mice (P < 0.001 in OVISE, and P = 0.038 in RMG-I) without severe body weight loss. Immunohistochemistry from the xenograft tumors showed that the combination treatment significantly reduced vascularity and cell proliferation, with an increase of apoptotic cell death. CONCLUSION: A combination therapy targeting the PI3K pathway and MDM2 might be a promising therapeutic strategy in CCOC.


Subject(s)
Ovarian Neoplasms/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Adenine/analogs & derivatives , Adenine/pharmacology , Adenocarcinoma, Clear Cell , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , DNA, Complementary/metabolism , Female , Heterografts , Imidazolines/pharmacology , Mice, Nude , Neoplasm Transplantation/physiology , Ovarian Neoplasms/metabolism , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/metabolism , RNA, Neoplasm/metabolism , Random Allocation
2.
Prostate ; 76(1): 97-113, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26417683

ABSTRACT

BACKGROUND: Previous results from our lab indicate a tumor suppressor role for the transmembrane protein with epidermal growth factor and two follistatin motifs 2 (TMEFF2) in prostate cancer (PCa). Here, we further characterize this role and uncover new functions for TMEFF2 in cancer and adult prostate regeneration. METHODS: The role of TMEFF2 was examined in PCa cells using Matrigel(TM) cultures and allograft models of PCa cells. In addition, we developed a transgenic mouse model that expresses TMEFF2 from a prostate specific promoter. Anatomical, histological, and metabolic characterizations of the transgenic mouse prostate were conducted. The effect of TMEFF2 in prostate regeneration was studied by analyzing branching morphogenesis in the TMEFF2-expressing mouse lobes and alterations in branching morphogenesis were correlated with the metabolomic profiles of the mouse lobes. The role of TMEFF2 in prostate tumorigenesis in whole animals was investigated by crossing the TMEFF2 transgenic mice with the TRAMP mouse model of PCa and analyzing the histopathological changes in the progeny. RESULTS: Ectopic expression of TMEFF2 impairs growth of PCa cells in Matrigel or allograft models. Surprisingly, while TMEFF2 expression in the TRAMP mouse did not have a significant effect on the glandular prostate epithelial lesions, the double TRAMP/TMEFF2 transgenic mice displayed an increased incidence of neuroendocrine type tumors. In addition, TMEFF2 promoted increased branching specifically in the dorsal lobe of the prostate suggesting a potential role in developmental processes. These results correlated with data indicating an alteration in the metabolic profile of the dorsal lobe of the transgenic TMEFF2 mice. CONCLUSIONS: Collectively, our results confirm the tumor suppressor role of TMEFF2 and suggest that ectopic expression of TMEFF2 in mouse prostate leads to additional lobe-specific effects in prostate regeneration and tumorigenesis. This points to a complex and multifunctional role for TMEFF2 during PCa progression.


Subject(s)
Adenocarcinoma , Carcinogenesis/metabolism , Membrane Proteins/metabolism , Neuroendocrine Tumors , Prostate , Prostatic Neoplasms , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Immunohistochemistry , Male , Mice , Mice, Transgenic , Neoplasm Transplantation/pathology , Neoplasm Transplantation/physiology , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Prostate/pathology , Prostate/physiology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Regeneration , Tumor Cells, Cultured
3.
Bull Exp Biol Med ; 158(4): 497-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25708334

ABSTRACT

Blood levels of vascular endothelial growth factor, the main marker of angiogenesis activity, and coagulation hemostasis were studied in CDF1 mice with transplanted P-388 lymphocytic leukemia. Blood levels of vascular endothelial growth factor increased by 168% in mice with tumors. The animals developed the hypercoagulation syndrome manifesting in shorter activated partial thromboplastin time and prothrombin time and development of hyperfibrinogenemia.


Subject(s)
Blood Coagulation Disorders/physiopathology , Leukemia P388/physiopathology , Neovascularization, Pathologic/physiopathology , Vascular Endothelial Growth Factor A/blood , Animals , Enzyme-Linked Immunosorbent Assay , Male , Mice , Neoplasm Transplantation/physiology , Neovascularization, Pathologic/blood , Partial Thromboplastin Time , Prothrombin Time , Statistics, Nonparametric
4.
Nat Commun ; 5: 5433, 2014 Nov 12.
Article in English | MEDLINE | ID: mdl-25388829

ABSTRACT

The parafibromin/hCdc73 is a component of the PAFc, which controls RNA polymerase II-mediated general transcription. In parathyroid carcinoma and familial autosomal dominant hyperparathyroidism-jaw tumour (HPT-JT), hCdc73 mutations are heavily implicated, yet the underlying mechanism of its carcinogenic action is poorly understood. Here we demonstrate that hCdc73 specifically controls messenger RNA stability of p53 and p53-mediated apoptosis. hCdc73 is associated with mature p53 mRNA in the cytoplasm and facilitates its degradation. Cytoplasmic hCdc73 physically interacts with eEF1Bγ and hSki8, and this interaction is required to bind and destabilize p53 mRNA. Furthermore, enhanced association of p53 mRNA with a cancer-driven hCdc73(K34Q) mutant was also observed. As a result, reduced p53 expression as well as enhanced cell proliferation was acquired in the hCdc73 (K34Q)-overexpressed cells. Altogether, our findings indicate that hCdc73 directly targets p53 mRNA to repress p53 expression, and aberrant regulation of this interaction may lead to tumour progression.


Subject(s)
Apoptosis/physiology , RNA, Messenger/physiology , Tumor Suppressor Protein p53/physiology , Tumor Suppressor Proteins/physiology , Animals , Carrier Proteins/physiology , Gene Expression Regulation, Neoplastic/physiology , Gene Silencing/physiology , Humans , Male , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation/physiology , RNA Stability/physiology
5.
J Neurooncol ; 106(1): 59-70, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21735115

ABSTRACT

Medulloblastoma is one of the leading causes of morbidity and mortality in pediatric cancer. Wnt-active tumors, an independent molecular subgroup in medulloblastoma, are characterized by a distinct pattern of genomic aberrations. We assessed the anticancer activity of cantharidin and norcantharidin against medulloblastoma, as cell lines in vitro and in athymic nude mice in vivo. Cantharidin and norcantharidin treatment impaired the growth of DAOY and UW228 medulloblastoma cells and promoted the loss of ß-catenin activation and the ß-catenin nuclearization linked to N-cadherin impairment in vitro. Intra-peritoneal administration of norcantharidin inhibited the growth of intra-cerebellum tumors in orthotopic xenograft nude mice. Analysis of the xenograft tissues revealed enhanced neuronal differentiation and reduced ß-catenin expression. Our findings suggest that norcantharidin has potential therapeutic applications in the treatment of medulloblastoma as a result of its ability to cross the blood-brain barrier and its impairment of Wnt-ß-catenin signaling.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Medulloblastoma/drug therapy , Wnt Proteins/physiology , beta Catenin/physiology , Animals , Apoptosis/physiology , Blood-Brain Barrier/physiology , Brain Neoplasms/pathology , Cell Cycle/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Flow Cytometry , Fluorescent Antibody Technique , G2 Phase/drug effects , Genes, Reporter , Indicators and Reagents , Luciferases/genetics , Medulloblastoma/pathology , Mice , Mice, SCID , Neoplasm Transplantation/physiology , Polymerase Chain Reaction , Protein Transport/physiology , Signal Transduction/physiology , Wnt Proteins/antagonists & inhibitors , beta Catenin/antagonists & inhibitors
6.
Differentiation ; 73(2-3): 88-98, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15811132

ABSTRACT

We investigated alpha1-antichymotrypsin (ACT) gene expression in xenograft tumors generated by two isogenic human breast cancer cell lines derived from the same parent, MDA-MB-435, which display opposite metastatic behaviors. Microarray and real-time PCR experiments showed an overexpression of this serine protease inhibitor in the metastatic tumors (M-4A4T) and its derived metastases (M4-Mets) compared with the weakly metastatic tumors (NM-2C5T), and its release into the blood was confirmed by western-blotting. However, functional assays in vivo using genetically engineered tumor cells demonstrated that ACT up-regulation was not, by itself, responsible for the metastatic phenotype. We also made observations that ACT gene regulation was sensitive to tumor-host interactions: inoculation of these lines into the mouse mammary gland greatly increased ACT production and accentuated the intrinsic difference observed when they are cultured in vitro. Sensitivity of tumor cells to their environment was further analyzed by in vitro experiments, which demonstrated that a purified ECM environment and soluble components from normal host mammary cells were both able to significantly promote ACT expression. In addition, we took advantage of the xenogeneic nature of the model to measure ACT expression by the host cells (mouse) and the tumor cells (human) within the neoplasm using species-specific primers in real-time PCR experiments. It was found that the presence of tumor cells, irrespective of their metastatic capabilities, induced local ACT production by host cells at the primary and secondary tumor sites. Thus, this work indicates that there is a specific association of ACT overexpression with the metastatic phenotype in our breast cancer metastasis model. Moreover, because of the xenogeneic nature of our system, we were able to provide evidence of tumor-host reciprocal regulation of ACT production.


Subject(s)
Breast Neoplasms/pathology , Transplantation, Heterologous/physiology , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred BALB C , Mice, SCID , Neoplasm Metastasis , Neoplasm Transplantation/physiology , Oligonucleotide Array Sequence Analysis , RNA, Neoplasm/genetics , RNA, Neoplasm/isolation & purification , Serpins/genetics
7.
Stem Cells ; 23(5): 699-706, 2005 May.
Article in English | MEDLINE | ID: mdl-15849177

ABSTRACT

Fibroblasts, which are widely distributed and play a key part in tissue fibrosis, are phenotypically and functionally heterogeneous. Recent studies reported that bone marrow can be a source of tissue fibroblast. In the study reported here, we investigated in vivo characterization of bone marrow-derived fibroblasts recruited into various fibrotic lesions. Mice were engrafted with bone marrow isolated from transgenic mice expressing green fluorescent protein (GFP), and fibrotic lesions were induced by cancer implantation (skin), excisional wounding (skin), and bleomycin administration (lung). A small population of GFP+ fibroblast was found even in nonfibrotic skin (8.7% +/- 4.6%) and lung (8.9% +/- 2.5%). The proportion of GFP+ fibroblasts was significantly increased after cancer implantation(59.7% +/- 16.3%) and excisional wounding (32.2% +/- 4.8%), whereas it was not elevated after bleomycin administration (7.1% +/- 2.4%). Almost all GFP+ fibroblasts in fibrotic lesions expressed type I collagen, suggesting that bone marrow-derived fibroblasts would contribute to tissue fibrosis. GFP+ fibroblasts expressed CD45, Thy-1, and alpha-smooth muscle actin at various proportions. Our results suggested that bone marrow-derived fibroblasts expressed several fibroblastic markers in vivo and could be efficiently recruited into fibrotic lesions in response to injurious stimuli; however, the degree of recruitment frequency might depend on the tissue microenvironment.


Subject(s)
Bone Marrow Cells/physiology , Bone Marrow Transplantation , Cell Lineage/physiology , Fibroblasts/physiology , Pulmonary Fibrosis/physiopathology , Animals , Antibiotics, Antineoplastic/toxicity , Bleomycin/toxicity , Bone Marrow Cells/cytology , Fibroblasts/cytology , Fibrosis/pathology , Fibrosis/physiopathology , Mice , Mice, Knockout , Neoplasm Transplantation/pathology , Neoplasm Transplantation/physiology , Neoplasms, Experimental/pathology , Neoplasms, Experimental/physiopathology , Pulmonary Fibrosis/chemically induced , Skin/pathology , Skin/physiopathology , Wound Healing/physiology
8.
Comb Chem High Throughput Screen ; 8(8): 789-99, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16464166

ABSTRACT

During the course of biosynthesis, processing and degradation of a peptide, many structurally related intermediate peptide products are generated. Human body fluids and tissues contain several thousand peptides that can be profiled by reversed-phase chromatography and subsequent MALDI-ToF-mass spectrometry. Correlation-Associated Peptide Networks (CAN) efficiently detect structural and biological relations of peptides, based on statistical analysis of peptide concentrations. We combined CAN with recognition of probable cleavage sites for peptidases and proteases in cerebrospinal fluid, resulting in a model able to predict the sequence of unknown peptides with high accuracy. On the basis of this approach, identification of peptide coordinates can be prioritized, and a rapid overview of the peptide content of a novel sample source can be obtained.


Subject(s)
Biomarkers, Tumor , Neoplasm Transplantation/physiology , Peptides/chemistry , Severe Combined Immunodeficiency/metabolism , Animals , Cell Line, Tumor , Female , Mice , Mice, SCID , Peptide Library , Peptides/genetics , Spectrometry, Mass, Electrospray Ionization , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
9.
J Biopharm Stat ; 14(4): 931-45, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15587973

ABSTRACT

In cancer drug development, demonstrated anticancer activity in animal models is an important step to bring a promising compound to clinic. Proper design and analysis of experiments using laboratory animals have received increasing attention recently. These experiments involve informatively censored longitudinal data with small samples. The problem is further complicated because of order constraints due to the intrinsic growth of control tumors without treatment. This article proposes a Bayesian hierarchical model to analyze informatively censored longitudinal data while accounting for the parameter constraints and providing valid small sample inference. We adopt a noniterative sampling approach, the inverse Bayes formulae (IBF) sampler, to generate independent posterior samples, which avoids convergence problems associated with Markov chain Monte-Carlo methods. To effectively deal with the restricted parameter problem, we use a linear transformation to simplify the constraints and exploit the IBF method to generate random samples from truncated multivariate normal distributions. Because diffuse priors are used, the posterior modes approximate the maximum likelihood estimates well, and the hierarchical model can be considered as an extended mixed-effects model. A real xenograft experiment on a new treatment is analyzed by using the proposed method.


Subject(s)
Antineoplastic Agents/therapeutic use , Dacarbazine/analogs & derivatives , Neoplasm Transplantation/physiology , Algorithms , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bayes Theorem , Brain Neoplasms/drug therapy , Camptothecin/therapeutic use , Dacarbazine/therapeutic use , Humans , Likelihood Functions , Longitudinal Studies , Markov Chains , Mice , Models, Statistical , Monte Carlo Method , Neuroblastoma/drug therapy , Predictive Value of Tests , Temozolomide , Transplantation, Heterologous
10.
J Biopharm Stat ; 14(4): 947-67, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15587974

ABSTRACT

In this article, we present methodology for making inferences about projected completors in the presence of attrition. The approach is motivated by a clinical trial that investigates a treatment for disability among individuals who sustain severe head injuries. Although most studies attempt to make inferences about the entire study population, our application poses important scientific questions targeting individuals who are likely to complete the study or to remain on protocol for a specified time period. We propose using measures of each individual's dropout inclination to identify projected completors and then building a stratified response model based on projected completion status. We present several prediction measures along with procedures for evaluating accuracy with respect to observed dropout. Estimation of model parameters proceeds using maximum likelihood and restricted maximum likelihood methods. We illustrate the utility of our proposed analysis by using the motivating disability data example.


Subject(s)
Antineoplastic Agents/therapeutic use , Dacarbazine/analogs & derivatives , Neoplasm Transplantation/physiology , Algorithms , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bayes Theorem , Brain Neoplasms/drug therapy , Camptothecin/therapeutic use , Dacarbazine/therapeutic use , Humans , Likelihood Functions , Longitudinal Studies , Markov Chains , Mice , Models, Statistical , Monte Carlo Method , Neuroblastoma/drug therapy , Predictive Value of Tests , Temozolomide , Transplantation, Heterologous
11.
J Exp Ther Oncol ; 4(2): 111-9, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15500006

ABSTRACT

Insulin-like growth factor receptor 1 (IGF-1R) plays a critical role in oncogenic transformation (1). IGF-1R is overexpressed in some tumors including breast, lung, cervical, and Wilms' tumors (2-6). Upon binding of IGF-I or IGF-II, IGF-1R, a tyrosine kinase, phosphorylates tyrosine residues on two major substrates, IRS-1 and Shc, which subsequently signal through the Ras/Raf and PI 3-kinase/AKT pathways (7). Extensive literature has shown that when the IGF-1R signaling pathway is blocked by antisense, dominant negative truncation or neutralizing antibodies, cellular transformation and tumor formation in mice is inhibited (8-18). Small molecule kinase inhibitors represent a valid approach to inhibit activity and downstream signalling of IGF-1R. To date, few potent and selective small molecule inhibitors of IGF-1R kinase activity have been reported. We expressed the tyrosine kinase domain of IGF-1R (IGF-1R/TK) in insect cells and subsequently purified the partially activated IGF-1R/TK. A compound library has been screened using a homogeneous time-resolved fluorescence (HTRF) assay. The hits generated by HTRF were then evaluated in a 33P ATP streptavidin-Flashplate assay (Flashplate). There was approximately 78% hit congruence between the two assay formats. One compound, C100, inhibited the IGF-1R kinase activity with an IC50 of 1 microM. C100 also inhibited IGF-1R autophosphorylation, AKT and MAPK activations in cells. This inhibitor provides a useful tool for studying IGF-1R in cells.


Subject(s)
Clinical Laboratory Techniques , Protein Kinase Inhibitors/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Breast Neoplasms , Cell Line, Tumor , Fluorescence , Humans , Neoplasm Transplantation/physiology , Protein-Tyrosine Kinases/antagonists & inhibitors , Signal Transduction/drug effects
12.
Mol Endocrinol ; 18(9): 2333-43, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15192074

ABSTRACT

We have investigated the molecular mechanisms involved in 17 beta-estradiol-induced angiogenic pathway. We show here that 17 beta-estradiol promoted a 6-fold increase in Jagged1 expression and an 8-fold increase in Notch1 expression by cDNA arrays in breast cancer MCF7 cells. Interestingly, Jagged1 was abrogated by incubation with the estrogen antagonist, ICI182,780. A similar up-regulation of both Notch1 receptor and Jagged1 ligand was found in endothelial cells. Additionally, imperfect estrogen-responsive elements were found in the 5' untranslated region of Notch1 and Jagged1 genes. Treatment with 17 beta-estradiol also led to an activation of Notch signaling in MCF7 cells expressing Notch1 reporter gene or by promoting Jagged1-induced Notch signaling in coculture assays. Inoculation of MCF7 cells in 17 beta-estradiol-treated nude mice resulted in up-regulation of Notch1 expression as well as increased number of tumor microvessels in comparison to placebo-treated mice. Notch1-expressing endothelial cell cultures formed cord-like structures on Matrigel in contrast to cells expressing a dominant-negative form of Notch1, emphasizing the relevance of Notch1 pathway in vessel assembly. Finally, Notch1-expressing MCF7 cells up-regulated hypoxia-inducible factor 1 alpha gene, a well-known angiogenic factor that clustered with Notch1 gene. This study implicates Notch signaling in the cross talk between 17 beta-estradiol and angiogenesis.


Subject(s)
Breast Neoplasms/blood supply , Estrogens/physiology , Neovascularization, Pathologic/genetics , Receptors, Cell Surface/metabolism , Signal Transduction , Transcription Factors/metabolism , Animals , Breast Neoplasms/metabolism , Calcium-Binding Proteins , Cell Line, Tumor , Estrogens/pharmacology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Intercellular Signaling Peptides and Proteins , Jagged-1 Protein , Membrane Proteins/genetics , Mice , Neoplasm Transplantation/physiology , Neovascularization, Pathologic/metabolism , Oligonucleotide Array Sequence Analysis , Receptor, Notch1 , Receptors, Cell Surface/genetics , Receptors, Cell Surface/physiology , Serrate-Jagged Proteins , Transcription Factors/genetics , Transcription Factors/physiology , Up-Regulation
13.
Stem Cells ; 22(2): 188-201, 2004.
Article in English | MEDLINE | ID: mdl-14990858

ABSTRACT

The aim of this study was to investigate factors influencing the engraftment potential of acute myeloid leukemia (AML) CD34+ cells in nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice. We examined the relationship between engraftment, CXCR4 expression on CD34+ and CD34+CD38- cells, and patient (Pt) clinical/laboratory characteristics in 44 samples from 11 Pts. Engraftment, evaluated by Southern blot and CD45 flow cytometric analyses, was observed in murine bone marrow of 6 of 11 Pt samples, ranging from 0.1% to 73.9% by Southern blot and from 0.1%-36.8% by flow cytometry. Poor Pt prognosis was inversely correlated with engraftment; the median overall survival was 95.9 weeks for Pts whose cells did not engraft and 26.1 weeks for those whose cells did engraft (p = 0.012, log-rank test). No other clinical/laboratory variable predicted engraftment. No correlation between the level of CXCR4 expression on AML cells and engraftment was observed. Cells with virtually absent CXCR4 expression were able to engraft, and cells from two Pts with high expression levels of CXCR4 did not engraft. Furthermore, anti-CXCR4 antibody failed to block the engraftment of AML cells into NOD/SCID mice. In conclusion, we demonstrated that CXCR4 is not critical for the engraftment of AML CD34+ cells in NOD/SCID mice. The model may, however, reflect the clinical course of the disease.


Subject(s)
Bone Marrow/metabolism , Graft Survival/physiology , Leukemia, Myeloid, Acute , Receptors, CXCR4/metabolism , Animals , Antigens, CD34/immunology , Flow Cytometry , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation/physiology
14.
Biomed Tech (Berl) ; 47 Suppl 1 Pt 1: 451-4, 2002.
Article in German | MEDLINE | ID: mdl-12451890

ABSTRACT

Due to the high solubility of molecular oxygen in perfluorocabons (PFC), this class of fluorinated compounds has gained wide-spread interest for its biomedical application as temporary blood substitutes and as radiosensitizers. Since the observation that the NMR spin-lattice relaxation times (T1) of some 19F PFC resonances are sensitive to oxygen tension (pO2), this paramagnetic effect has been used to non-invasively probe pO2 in vivo. In this study, combined 19F/1H NMR image data of Copenhagen rats after PFC application were evaluated with the software package MATLAB. The analysis of the 19F NMR data resulted in image matrices with calculated T1 values in each pixel. By using a calibration curve, the corresponding pO2 values were computed. Color overlays of pO2 contour lines on T1-weighted 1H images show a good anatomical-functional correspondence.


Subject(s)
Fluorocarbons , Image Processing, Computer-Assisted , Magnetic Resonance Spectroscopy , Oxygen Consumption/physiology , Prostatic Neoplasms/physiopathology , Animals , Artifacts , Male , Neoplasm Transplantation/physiology , Rats , Rats, Inbred Strains , Sensitivity and Specificity
15.
Invest New Drugs ; 20(3): 241-51, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12201487

ABSTRACT

In cell culture, the compound 317615 2HCl, a potent inhibitor of VEGF-stimulated HUVEC proliferation, was not very effective against MX-1 breast cancer cells (IC50= 8.1 microM) or SKOV-3 ovarian carcinoma cells (IC50 = 9.5 microM). Exposure to combinations of paclitaxel or carboplatin and 317615 x 2HCl with MX-1 cells in culture resulted in cell survival that reflected primarily additivity of the two agents. Exposure of SKOV-3 cells to paclitaxel or carboplatin along with 317615 2HCl resulted in cell survivals that reflected additivity of 317615 x 2HCl with paclitaxel and greater-than-additive cytotoxicity with carboplatin. Administration of 317615 x 2HCI orally twice daily to nude mice bearing subcutaneous MX-1 tumors or SKOV-3 tumors resulted in a decreased number of intratumoral vessels as determined by CD31 and CD105 staining with decreases of 35% and 43% in MX-1 tumors and 60% and 75% in SKOV-3 tumors, respectively. 317615 x 2HCl was an active antitumor agent against the MX-1 xenograft and increased the tumor growth delay produced by paclitaxel by 1.7-fold and the tumor growth delay produced by carboplatin by 3.8-fold. Administration of 317615 x 2HCl also increased the tumor growth delay produced by fractionated radiation therapy in the MX-1 tumor. Treatment with 317615 x 2HCl alone increased the lifespan of animals bearing intraperitoneal SKOV-3 xenografts by 1.9 fold compared with untreated control animals. The combination of paclitaxel and 317615 x 2HCl resulted in 100% 120-day survival of SKOV-3 bearing animals. Administration of 317615 x 2HCl along with carboplatin to animals bearing the SKOV-3 tumor produced a 1.8-fold increase in lifespan compared with carboplatin alone. 317615 x 2HCl is a promising new antiangiogenic agent that is in early phase clinical testing.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Breast Neoplasms/drug therapy , Enzyme Inhibitors/therapeutic use , Ovarian Neoplasms/drug therapy , Protein Kinase C/antagonists & inhibitors , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Breast Neoplasms/blood supply , Breast Neoplasms/radiotherapy , Carboplatin/therapeutic use , Cell Survival/drug effects , Female , Humans , Mice , Mice, Nude , Neoplasm Transplantation/physiology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Organic Chemicals , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/radiotherapy , Paclitaxel/therapeutic use , Protein Kinase C beta , Regional Blood Flow/drug effects , Transplantation, Heterologous
16.
Med Pediatr Oncol ; 36(1): 154-6, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11464872

ABSTRACT

BACKGROUND: The insulin-like growth factors (IGFs) are involved in the growth and differentiation of neuroblastoma cells. In all biological fluids, they are non-covalently bound to high-affinity binding proteins (IGFBP-1 to -6) which modulate their bioavailability. We previously showed that IGFBP-6 expression is linked to the arrest of growth in neuroblastoma cells, whereas IGFBP-2 is associated with proliferation. PROCEDURE: To study the role of IGFBP-6 in cell growth, we stably IGR-N-91 neuroblastoma cells with a plasmid containing sequences coding for IGFBP-6 under the control of the cytomegalovirus (CMV) promoter. RESULTS: The incidence and size of tumors generated by injecting IGFBP-6-expressing cells into nude mice were reduced by factors of 2 and 5, respectively, as compared with those generated by injection by control cells. Northern blot analyses if xenografts revealed weaker expression of IGF-II, type 2 IGF receptor and IGFBP-2 mRNAs in IGFBP-6-expressing cthan in control xenografts. IGFBP-6 may therefore reduce the expression of IGF-II (which induces tumour development) at a transcriptional level. Conversely, containing IGFBP-2 cDNA under the control of CMV promoter grew three to four times as fast as normal control xenografts. Northern blot analyses revealed weaker expression of intact IGFBP-3 and IGFBP-1 in IGFBP-2-expressing than in control xenografts. CONCLUSIONS: IGFBP-1 and intact IGFBP-3 expression both enhance IGF bioavailability which promotes tumour growth. Although the mechanisms of action of IGFBP-2 and IGFBP-6 remain to be elucidated, an inverse relationship appears to exist between the two binding proteins, IGFBP-2 being involved in proliferation and IGFBP-6 in its arrest.


Subject(s)
Insulin-Like Growth Factor Binding Proteins/physiology , Neoplasm Transplantation/physiology , Neuroblastoma/pathology , Transplantation, Heterologous/physiology , Animals , Biological Availability , Cytomegalovirus/genetics , Genes, Synthetic , Graft Survival , Humans , Insulin-Like Growth Factor Binding Protein 2/genetics , Insulin-Like Growth Factor Binding Protein 2/physiology , Insulin-Like Growth Factor Binding Protein 6/genetics , Insulin-Like Growth Factor Binding Protein 6/physiology , Insulin-Like Growth Factor I/physiology , Insulin-Like Growth Factor II/physiology , KB Cells/metabolism , KB Cells/transplantation , Mice , Mice, Nude , Mitosis , Promoter Regions, Genetic , Recombinant Fusion Proteins/physiology , Transfection , Transgenes , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/transplantation
17.
Cell Transplant ; 10(2): 175-81, 2001.
Article in English | MEDLINE | ID: mdl-11332632

ABSTRACT

In a model of transplantation rejection, we have tested whether a graft manipulated to secrete immunomodulators could protect itself from immune destruction. An insulinoma cell line having the NOD genotype but also expressing the neoantigen, SV40 T antigen, was transfected with CTLA4Ig or LFA3Ig to block signals in the co-stimulatory/adhesion pathways. This neoantigen is potent at inducing graft rejection. Secretion of CTLA4Ig and LFA3Ig by transfectants promoted survival of the insulinoma graft in young NOD mice. In immunodeficient mice, cell growth was similar for all transfectants. However, in immunocompetent NOD mice the survival/growth of test grafts was significantly better than that of the controls. Graft survival was enhanced additively, when the two test transfectants were cotransplanted. Endowing the graft the ability to secrete immunomodulators that block individual co-stimulatory/adhesion signals can contribute to transplantation success. Blockade of two signals (CD2 and CD28) in these pathways enhances this success.


Subject(s)
B7-1 Antigen/immunology , CD2 Antigens/immunology , Graft Survival/physiology , Immunoconjugates , Neoplasm Transplantation/physiology , Abatacept , Alefacept , Animals , Antigens, CD , Antigens, Differentiation/genetics , Antigens, Differentiation/immunology , Antigens, Polyomavirus Transforming/genetics , CTLA-4 Antigen , Immunosuppression Therapy/methods , Immunosuppressive Agents , Insulin/metabolism , Insulin Secretion , Insulinoma/metabolism , Insulinoma/pathology , Mice , Mice, Inbred NOD , Neoplasm Transplantation/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Recombinant Fusion Proteins/immunology , Transfection , Tumor Cells, Cultured
18.
J Pharmacol Exp Ther ; 291(3): 1269-75, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10565851

ABSTRACT

We previously demonstrated that sustained depletion of methylguanine DNA methyltransferase (MGMT) activity is required for optimal reversal of chloroethylnitrosourea resistance in tumor cells. The purpose of this study was to design O(6)-benzylguanine (BG) treatments that deplete MGMT activity in tumor cells and xenograft tumors in a prolonged manner. When SF767 cells were treated with a bolus dose of BG (25 microM for 1 h), >95% of MGMT activity was depleted but 33% of the activity recovered within 24 h. In contrast, MGMT activity was completely depleted for 24 h when cells were pretreated with a low dose of BG (2.5 microM) for 24 h, followed by the bolus dose and same low-dose treatment for 24 h. This combination regimen of pre- and post-treatments with a bolus dose sensitized cells N,N'-bis(2-chloroethyl)-N-nitrosourea in vitro by approximately 2-fold more than the bolus dose alone. Similar BG treatment with Alzet micro-osmotic pumps produced sustained inhibition of MGMT activity in vivo. In xenograft SF767 tumors, low-dose pre- and post-treatments (8 mg/kg over 24 h) combined with an i.p. bolus dose (80 mg/kg) of BG inhibited >95% of MGMT activity for 24 h after the bolus. The bolus dose alone did not deplete MGMT for 24 h. These results demonstrate that combination low-dose and bolus BG treatment is superior to the bolus dose alone in depleting MGMT activity in a sustained manner in vitro and in vivo. When combined with N,N'-bis(2-chloroethyl)-N-nitrosourea treatment, this BG regimen also should also produce greater antitumor activity than the single bolus dose evaluated clinically.


Subject(s)
Enzyme Inhibitors/pharmacology , Guanine/analogs & derivatives , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , Animals , Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/pathology , Carmustine/pharmacology , Female , Glioma/pathology , Guanine/pharmacology , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation/physiology , Tumor Cells, Cultured
19.
Neuroreport ; 9(12): 2837-42, 1998 Aug 24.
Article in English | MEDLINE | ID: mdl-9760130

ABSTRACT

We recently reported behavioral improvements following intrastriatal transplantation of cryopreserved cultured human neuroteratocarcinoma-derived cells (hNT neurons) in rats with cerebral ischemia induced by occlusion of the middle cerebral artery. In the present study, the viability and survival of hNT neurons were evaluated immediately prior to the transplantation surgery and at 3 months post-transplantation in ischemic rats. Cryopreserved hNT neurons were routinely thawed, and trypan blue exclusion viability counts revealed 52-95% viable hNT neurons before transplantation. Monthly behavioral tests, starting at 1 month and extending to 3 months post-transplantation, revealed that ischemic animals that were intrastriatally transplanted with hNT neurons (approximately 40000) and treated with an immunosuppressive drug displayed normalization of asymmetrical motor behavior compared with ischemic animals that received medium alone. Within-subject comparisons of cell viability and subsequent behavioral changes revealed that a high cell viability just prior to transplantation surgery correlated highly with a robust and sustained functional improvement in the transplant recipient. Furthermore, histological analysis of grafted brains revealed a positive correlation between number of surviving hNT neurons and degree of functional recovery. In concert with similar reports on fetal tissue transplantation, we conclude that high cell viability is an important criterion for successful transplantation of cryopreserved neurons derived from cell lines to enhance graft-induced functional effects.


Subject(s)
Brain Ischemia/pathology , Brain Tissue Transplantation/physiology , Cell Transplantation/physiology , Neoplasm Transplantation/physiology , Neurons/physiology , Neurons/transplantation , Animals , Brain Neoplasms/physiopathology , Cell Survival/physiology , Humans , Male , Neostriatum/pathology , Rats , Rats, Sprague-Dawley , Teratoma/physiopathology , Tumor Cells, Cultured
20.
J Cardiovasc Pharmacol ; 31 Suppl 1: S531-3, 1998.
Article in English | MEDLINE | ID: mdl-9595534

ABSTRACT

In cancer chemotherapy, selective enhancement of drug delivery to tumor tissue is essentially important for increase of chemotherapeutic effects. An attenuated vasoconstrictive response to angiotensin II (Ang II) in tumors and a marked increase in tumor blood flow were observed compared with normal tissues during systemic hypertension induced by Ang II infusion. The phenomenon was absent when hypertension was provoked by endothelin-1 (ET-1). We assessed this response to characterize ET receptor and Ang II receptor density and affinity in normal and tumor tissues. The tumor cell line LY80 was transplanted to the skin in nude rats. Four weeks later the rats were sacrificed. [125I] ET-1 and [125I Sar1, Ile8]-Ang II were used to map the receptors for ET and Ang II in rat tissues using computerized in vitro autoradiography. A moderately high density of ET receptors, (ETB > ETA) was found in tumors. The Ang II receptors were markedly reduced in tumor tissues without changes in the affinity. These results suggest that the decrease in Ang II receptors but not ET receptors in tumors may explain the hemodynamic effect of Ang II-induced hypertension and ET-induced hypertension on tumor blood flow.


Subject(s)
Angiotensin II/metabolism , Neoplasms, Experimental/metabolism , Receptors, Angiotensin/metabolism , Receptors, Endothelin/metabolism , Animals , Autoradiography , Cell Transplantation/physiology , Humans , Male , Neoplasm Transplantation/physiology , Rats , Transplantation, Heterologous/physiology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...